US20130157924A1 - Reducing transmission of sexually transmitted infections - Google Patents

Reducing transmission of sexually transmitted infections Download PDF

Info

Publication number
US20130157924A1
US20130157924A1 US13/643,026 US201113643026A US2013157924A1 US 20130157924 A1 US20130157924 A1 US 20130157924A1 US 201113643026 A US201113643026 A US 201113643026A US 2013157924 A1 US2013157924 A1 US 2013157924A1
Authority
US
United States
Prior art keywords
substituted
unsubstituted
sevi
bta
infection
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/643,026
Inventor
Stephen Dewhurst
Bradley Nilsson
Joanna Olsen
Jerry Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
University of Rochester
Original Assignee
University of California
University of Rochester
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of California, University of Rochester filed Critical University of California
Priority to US13/643,026 priority Critical patent/US20130157924A1/en
Publication of US20130157924A1 publication Critical patent/US20130157924A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF ROCHESTER
Assigned to UNIVERSITY OF ROCHESTER reassignment UNIVERSITY OF ROCHESTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DEWHURST, STEPHEN, NILSSON, BRADLEY, OLSEN, JOANNA
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: YANG, JERRY
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF ROCHESTER
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • A61K47/48238
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals

Definitions

  • HIV human immunodeficiency virus
  • HIV Acquired immunodeficiency syndrome
  • AIDS is a collection of symptoms and infections resulting from the specific damage to the immune system caused by HIV.
  • HIV human immunodeficiency virus
  • the methods comprise administering to a subject with or at risk of acquiring a sexually transmitted infection a semen-derived enhancer of viral infection (SEVI)-binding agent comprising a compound described herein, including, e.g., BTA-EG 4 and BTA-EG 6 .
  • SEVI-binding small molecule can, for example, comprise a hydrophobic molecule that incorporates into or binds the SEVI-fibrils or an anionic polypeptide supramolecular assembly.
  • the methods can further comprise administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
  • compositions comprising a first agent, which is a semen-derived enhancer of viral infection (SEVI)-binding agent or small molecule (e.g., a hydrophobic molecule that incorporates into ro binds the SEVI-fibrils or an anionic polypeptide supramolecular assembly) as described herein, and a second selected from the group consisting of an anti-viral, an anti-bacterial, and an anti-fungal agent.
  • SEVI semen-derived enhancer of viral infection
  • FIGS. 1A and 1B are graphs showing semen-derived amyloid fibrils referred to as SEVI (semen-derived enhancer of virus infection) stimulates inflammatory cytokine production by primary human macrophages.
  • FIG. 1A is a graph showing IL-1 ⁇ , levels and
  • FIG. 1B is a graph showing TNFI levels in primary human macrophages treated with SEVI or mock treated.
  • FIGS. 2A and 2B show a schematic of a potential mechanism of a microbicide against SEVI ( FIG. 2A ) and SEVI-binding molecules ( 2 B).
  • FIGS. 3A and 3B show that prostatic acid phosphatase (PAP) 248-286 forms fibrils.
  • PAP248-286 (10 mg/ml in PBS) was agitated at 37° C. and 14,000 RPM.
  • FIG. 3A is a graph showing the results of samples collected at 0 hours, 24 hours, 48 hours, 72 hours, and 96 hours that were subjected to Thioflavin T analysis.
  • FIG. 3B shows images of SEVI-fibrils visualized by electron microscopy. Samples were collected at 72 hours.
  • FIGS. 4A and 4B show that SEVI fibrils enhance HIV-1 infection in vitro. 5 ⁇ 10 4 CEM 5.25 cells were exposed to infectious HIV-1 (pNL43; 1.2 ng of virus, as determined by p24 ELISA assay) for 2 hours in the presence (10 or 25 ⁇ g/mL) or absence of SEVI.
  • FIG. 4A is a graph showing luciferase activity measured in cell lysates at 48 hours post infection.
  • FIG. 4B shows fluorescence microscopy images of GFP at 48 hours.
  • FIGS. 5A and 5B show that the Thioflavin-T analogs BTA-EG 4 and BTA-EG 6 inhibit SEVI mediated enhancement of HIV infection.
  • CEM 5.25 cells were exposed to infectious HIV-1 (IIIB) for 2 hours in the absence or presence (25 ⁇ g/mL) of SEVI fibrils; BTA-EG 4 ( FIG. 5A ) and EG 6 ( FIG. 5B ) was added at concentrations of 5.5, 11 and 16.5 ⁇ g/mL. Luciferase activity was measured in cell lysates 72 hrs post-infection. * indicates p value ⁇ 0.05.
  • FIGS. 6A and 6B show BTA-EG 4 and BTA-EG 6 inhibit semen mediated enhancement of HIV Infection.
  • HIV-1 IIIB virions were preincubated with 50% semen, with or without increasing concentrations of BTA-EG 4 ( FIG. 6A ) and BTA-EG 6 ( FIG. 6B ). After 10 minutes these stocks were diluted 15 fold into CEM 5.25 cells. Cells were washed after 1 hour and luciferase expression was measured at 48 hours to quantify the extent of infection. * indicates p value ⁇ 0.05.
  • FIGS. 7A and 7B show BTA-EG 4 and BTA-EG 6 decrease SEVI enhanced binding of HIV to target cells.
  • HIV-1 (IIIB) virions were pretreated with 10 ug/mL SEVI and added to Jurkat cells with or without increasing concentrations of BTA-EG 4 ( FIG. 7A ) or BTA-EG 6 ( FIG. 7B ). After 90 minutes, cells were washed to remove any unbound virus and bound virions were detected using a p24 ELISA.
  • FIG. 8 shows fluorescence polarization analysis of heparin binding to SEVI fibrils.
  • FIGS. 9A and 9B show that fluorescence polarization detects binding of BTA-EG 4 ( FIG. 9A ) and BTA-EG 6 ( FIG. 9B ) to SEVI fibrils.
  • 100 ug/mL of SEVI was mixed with 16 ug/mL FITC-heparin in varying concentrations of BTA-EG 4 or BTA-EG 6 .
  • Samples were incubated 1 hour at room temperature and polarized fluorescence intensities were measured.
  • FIGS. 10A and 10B show BTA-EG 4 and EG 6 are not toxic to cervical epithelial Cells.
  • the cervical epithelial cell lines A2En (endocervical) ( FIG. 10A ), 3EC1 and SiHa ( FIG. 10B ) were treated for 12 hours with BTA-EG 4 and BTA-EG 6 at concentrations up to 10 times greater than the inhibitory concentration. At 12 hours, viability was measured with Alamar Blue.
  • FIGS. 11A and 11B show BTA-EG 4 and BTA-EG 6 do not induce cytokine production in cervical epithelial cells.
  • A2En, 3EC1 and SiHa Cells were treated with BTA-EG4 or BTA-EG6 at varying concentrations for 6 hours. At 6 hours, supernatants were collected and cytokine production (IL-1 ⁇ ( FIG. 11A ); Mip3 ⁇ ( FIG. 11B ); and TNF- ⁇ ) was determined by ELISA. Representative results from Siha cells are shown.
  • FIG. 12 shows that the thioflavin-T analog BTA-EG 6 binds SEVI fibrils.
  • FIG. 12A shows the chemical structure of ThT and BTA-EG 6 .
  • FIG. 12B shows that BTA-EG 6 binds SEVI fibrils as measured by fluorescence polarization.
  • 100 ⁇ g/ml SEVI was mixed with 16 ⁇ g/ml FITC-heparin in varying concentrations of BTA-EG 6 ranging from 0 to 200 ⁇ g/ml. Samples were incubated 1 h at room temperature, and polarized fluorescence intensities were measured. Decreased millipolarization units (mP) indicate a displacement of FITC-heparin from SEVI fibrils due to BTA binding.
  • FIG. 12 shows that the thioflavin-T analog BTA-EG 6 binds SEVI fibrils.
  • FIG. 12A shows the chemical structure of ThT and BTA-EG 6 .
  • FIG. 12B shows that BTA-EG 6
  • FIG. 12C shows binding of BTA-EG 6 to SEVI fibrils as determined by a centrifugation assay. Briefly, various concentrations of BTA-EG 6 in PBS were incubated overnight at room temperature in the presence or absence of SEVI fibrils. After equilibration, each solution was centrifuged, and the supernatants were separated from the pelleted fibrils. The fluorescence of BTA-EG 6 was determined from the resuspended pellets in PBS solution. Error bars represent ⁇ S.D. of duplicate measurements.
  • FIG. 12D shows that BTA-EG 6 does not affect the stability of SEVI fibrils. Preformed SEVI fibrils were incubated with increasing concentrations of BTA-EG 6 for 3 h. Fibril stability was measured by ThT fluorescence.
  • FIG. 12E shows that BTA-EG 6 binding to SEVI inhibits the interaction of SEVI fibrils with the cell surface.
  • Jurkat T cells were incubated with SEVI-biotin for 1 h in the presence or absence of 5.5 ⁇ g/ml (low) or 27 ⁇ g/ml (high) BTA-EG 6 .
  • Surface-bound fibrils were detected with SA-FITC and measured by flow cytometry. Results are summarized in Table 1 and are representative of three experiments that were performed with similar results.
  • FIG. 13 shows that BTA-EG 6 inhibits SEVI-mediated enhancement of HIV-1 infection.
  • HIV-1 IIIB virions were preincubated with increasing concentrations of BTA-EG 6 (0, 5.5, 11, and 22.5 ⁇ g/ml) and with or without SEVI (15 ⁇ g/ml) as indicated. The samples were then added to CEM-M7 cells. Cells were washed at 2 h, and infection was assayed at 48 h by measuring Tat-driven luciferase expression. Results shown are average values ⁇ S.D. of triplicate measurements from one of four independent experiments that yielded equivalent results. * indicates p ⁇ 0.05 when compared with control cells exposed to HIV-1 IIIB +SEVI alone by ANOVA with Tukey's post test.
  • FIG. 13B is a zoom in of panel A to show data for cells treated with HIV-IIIB virions with and without increasing concentrations of BTA-EG 6 , in the absence of SEVI. BTA-EG 6 had no effect on the infectivity of HIV alone; concentrations of BTA-EG 6 are noted above for panel A.
  • FIG. 13C shows the results of CEM-M7 cells infected with HIV-1 ADA , as in panel A.
  • FIG. 13D shows that CEM-M7 cells were infected with HIV-1 ADA +SEVI with concentrations of BTA-EG 6 ranging from 0.4 to 50 ⁇ g/ml.
  • FIG. 14 shows that BTA-EG 6 inhibits semen-mediated enhancement of HIV-1 infectivity.
  • FIG. 14A shows HIV-1 IIIB virions were preincubated with 50% pooled human semen, with or without increasing concentrations of BTA-EG 6 (5.5, 11, and 22.5 ⁇ g/ml). After 10 min, these stocks were diluted 15-fold into CEM-M7 cells. Cells were washed after 1 h, and luciferase expression was measured at 48 h to quantify the extent of infection. Results shown are average values ⁇ S.D. of triplicate measurements from one of three independent experiments that yielded equivalent results. * indicates p ⁇ 0.05 when compared with control cells exposed to HIV-1 IIIB +semen alone, by ANOVA with Tukey's post test.
  • FIG. 14B cells were treated as above but with HIV-1 ADA and a 50% concentration of an individual semen sample. *, p ⁇ 0.05 when compared with control cells exposed to HIV-1 ADA +semen alone, by ANOVA with Tukey's post test.
  • FIG. 15 shows that BTA-EG 6 inhibits SEVI-mediated attachment of HIV-1 to the cell surface.
  • HIV-1 IIIB virions were pretreated with or without 10 ⁇ g/ml SEVI and added to Jurkat cells with or without increasing concentrations of BTA-EG 6 (5.5, 11, and 22.5 ⁇ g/ml). After 90 min, cells were washed to remove any unbound virus, and bound virions were detected using a p24 ELISA.
  • FIGS. 15C A2En cells were incubated with HIV-1 ADA in the presence or absence of 22.5_g/ml BTA-EG 6 (* indicates p ⁇ 0.01 for cells treated with SEVI plus 22.5 ⁇ g/ml BTA-EG 6 versus cells treated with SEVI alone; ANOVA with Tukey's post test).
  • FIGS. 15A-C all results shown are average values ⁇ S.D. of triplicate measurements from one of three independent experiments that yielded equivalent results.
  • FIG. 15D A2En cells were treated with HIV-1BaL and 15 ⁇ g/ml SEVI with or without increasing concentrations of BTA-EG 6 (5.5, 11, and 22.5 ⁇ g/ml).
  • FIG. 16 shows that BTA-EG 6 is not toxic to cervical cells.
  • the cervical endothelial cell lines A2En endocervical
  • 3EC1 ectocervical
  • SiHa were treated for 12 h with BTA-EG 6 at concentrations up to 10 times greater than the IC 50 .
  • Control cultures were treated with nonoxynol-9 (non-9) at 0.1% final concentration as a positive control for induction of cell death.
  • viability was measured by resazurin cytotoxicity assay (AlamarBlue assay). Representative results from A2En cells are shown; results from 3EC1 and SiHa cells were very similar.
  • BTA-EG 6 does not induce inflammatory chemokine production in cervical epithelial cells.
  • A2En, 3EC1, and SiHa Cells were treated with BTA-EG 6 at varying concentrations for 6 h; control cultures were treated with a well defined TLR2/6 agonist, FSL1 (a synthetic diacylated lipoprotein derived from M. salivarium ) at 0.1 ⁇ g/ml final concentration as a positive control for chemokine induction.
  • FSL1 a synthetic diacylated lipoprotein derived from M. salivarium
  • FIGS. 17A and B show levels of bound virons using an HIV-1 p24 antigen capture assay with HIV-1 IIIB virions pretreated with 15 ⁇ g/ml SEVI and added to 5 ⁇ 10 4 A2En cells (immortalized primary human endocervical cells) (A) or to Jurkat T cells (a CD4+ human T cell line) (B) in the presence or absence of test compounds (at a final concentration of 25 ⁇ M).
  • A2En cells immortalized primary human endocervical cells
  • Jurkat T cells a CD4+ human T cell line
  • FIG. 18A shows a schematic of binding of an amyloid-binding ligand, like benzothiazole aniline (BTA), in monomeric (left panel) or oligomeric (right panel) form.
  • FIG. 18B shows the structure of a benzothiazole aniline (BTA)-based monomer (1), dimer (2), trimer (3), tetramer (4), and pentamer (5).
  • BTA benzothiazole aniline
  • the structure of BTA moiety is given and is represented as simple red ovals in molecules 1-5 for clarity.
  • sexual transmission of HIV can occur when HIV-containing secretions, e.g., seminal or vaginal fluid, of one partner come into contact with the genital, oral, or rectal mucous membranes of another.
  • HIV-containing secretions e.g., seminal or vaginal fluid
  • the epithelial cells of the mucous membranes act, at least in part, as a barrier to viral penetration. HIV can cross the epithelial barrier either by capture by intra-epithelial dendritic cells that convey the virus to target cells deeper in the mucosa or through regions of damaged epithelium resulting from traumatic injury or lesions caused by sexually transmitted diseases.
  • the virus Once the virus has breached the epithelial membrane, the infection spreads among cells of the immune system, including, for example, CD4+ T cells, macrophages and dendritic cells. Ultimately, the virus disseminates via the lymphatic system and the blood to spleen, brain, liver, and lungs.
  • the efficiency of sexual transmission of HIV depends on many factors, including, for example, host factors in both the transmitting partner and the recipient.
  • Seminal fluid contains a number of factors, for example, semen fibrils, amines such as spermine, spermidine, putrescine and cadavarine, as well as nutrients and enzymes that protect the virus from the acidic environment of the vaginal tract and that enhance sexual transmission of HIV.
  • Cationic polymers enhance retrovirus transduction by neutralizing the electrostatic repulsion between the virus and cell surface and allowing many virus particles to aggregate onto a single surface enhancing the effective multiplicity of infection.
  • semen fibrils e.g., prostatic acid phosphatase (PAP) fibrils
  • PAP prostatic acid phosphatase
  • PAP has been shown to be a safe vaccine antigen in the context of immunization for prostate cancer. Thus, immunization with short linear peptides derived from PAP is safe.
  • An amyloid-binding small molecule is an efficient inhibitor of SEVI- and semen-mediated enhancement of HIV infectivity.
  • BTA-EG 6 binds to the SEVI fibrils and interferes with their ability to enhance HIV infectivity. Importantly, BTA-EG 6 did not have any direct inhibitory effects on the infectivity of HIV-1 alone.
  • the methods optionally, comprise identifying a subject with or at risk of developing a sexually transmitted infection and administering to the subject a semen-derived enhancer of viral infection (SEVI)-binding agent, wherein the agent comprises a compound represented by Formula I:
  • the agent can, for example, bind and prevent the ability of SEVI-fibrils or prefibrillar forms of SEVI from enhancing a sexually transmitted infection in the subject.
  • the methods further comprise administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , and R 8 are each independently selected from hydrogen, halogen, hydroxyl, trifluoromethyl, substituted or unsubstituted thio, substituted or unsubstituted alkoxyl, substituted or unsubstituted aryloxyl, substituted or unsubstituted amino, substituted or unsubstituted C 1-12 alkyl, substituted or unsubstituted C 2-12 alkenyl, substituted or unsubstituted C 2-12 alkynyl, substituted or unsubstituted C 1-12 heteroalkyl, substituted or unsubstituted C 2-12 heteroalkenyl, substituted or unsubstituted C 2-12 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted, substituted
  • R 9 and R 10 are each independently selected from hydrogen, substituted or unsubstituted C 1-20 alkyl, substituted or unsubstituted C 2-20 alkenyl, substituted or unsubstituted C 2-20 alkynyl, substituted or unsubstituted C 1-20 heteroalkyl, substituted or unsubstituted C 2-20 heteroalkenyl, substituted or unsubstituted C 2-20 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 9 and R 10 can combine with the adjacent N to form a heterocycle.
  • R 10 is hydrogen.
  • R 9 is the Structure I-A:
  • n is an integer from 0 to 20. In some examples of Structure I-A, n is 4 or 6.
  • adjacent R groups on the phenyl ring i.e., R 1 , R 2 , R 3 , and R 4 ; R 5 and R 6 ; and R 7 and R 8
  • R 1 , R 2 , R 3 , and R 4 ; R 5 and R 6 ; and R 7 and R 8 can be combined to form substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, substituted or unsubstituted cycloalkynyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, or substituted or unsubstituted heterocycloalkynyl groups.
  • R 1 can be a formamide group and R 2 can be an ethylene group that combine to form a pyridinone group.
  • Other adjacent R groups include the combinations of R 2 and R 3 , R 3 and R 4 , R 5 and R 6 , and R 7 and R 8 .
  • alkyl, alkenyl, and alkynyl include straight- and branched-chain monovalent substituents. Examples include methyl, ethyl, isobutyl, 3-butynyl, and the like. Ranges of these groups useful with the compounds and methods described herein include C 1 -C 20 alkyl, C 2 -C 20 alkenyl, and C 2 -C 20 alkynyl.
  • Additional ranges of these groups useful with the compounds and methods described herein include C 1 -C 12 alkyl, C 2 -C 12 alkenyl, C 2 -C 12 alkynyl, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, and C 2 -C 4 alkynyl.
  • Heteroalkyl, heteroalkenyl, and heteroalkynyl are defined similarly as alkyl, alkenyl, and alkynyl, but can contain O, S, or N heteroatoms or combinations thereof within the backbone. Ranges of these groups useful with the compounds and methods described herein include C 1 -C 20 heteroalkyl, C 2 -C 20 heteroalkenyl, and C 2 -C 20 heteroalkynyl.
  • Additional ranges of these groups useful with the compounds and methods described herein include C 1 -C 12 heteroalkyl, C 2 -C 12 heteroalkenyl, C 2 -C 12 heteroalkynyl, C 1 -C 6 heteroalkyl, C 2 -C 6 heteroalkenyl, C 2 -C 6 heteroalkynyl, C 1 -C 4 heteroalkyl, C 2 -C 4 heteroalkenyl, and C 2 -C 4 heteroalkynyl.
  • cycloalkyl, cycloalkenyl, and cycloalkynyl include cyclic alkyl groups having a single cyclic ring or multiple condensed rings. Examples include cyclohexyl, cyclopentylethyl, and adamantanyl. Ranges of these groups useful with the compounds and methods described herein include C 3 -C 20 cycloalkyl, C 3 -C 20 cycloalkenyl, and C 3 -C 20 cycloalkynyl.
  • Additional ranges of these groups useful with the compounds and methods described herein include C 5 -C 12 cycloalkyl, C 5 -C 12 cycloalkenyl, C 5 -C 12 cycloalkynyl, C 5 -C 6 cycloalkyl, C 5 -C 6 cycloalkenyl, and C 5 -C 6 cycloalkynyl.
  • heterocycloalkyl, heterocycloalkenyl, and heterocycloalkynyl are defined similarly as cycloalkyl, cycloalkenyl, and cycloalkynyl, but can contain O, S, or N heteroatoms or combinations thereof within the cyclic backbone. Ranges of these groups useful with the compounds and methods described herein include C 3 -C 20 heterocycloalkyl, C 3 -C 20 heterocycloalkenyl, and C 3 -C 20 heterocycloalkynyl.
  • Additional ranges of these groups useful with the compounds and methods described herein include C 5 -C 12 heterocycloalkyl, C 5 -C 12 heterocycloalkenyl, C 5 -C 12 heterocycloalkynyl, C 5 -C 6 heterocycloalkyl, C 5 -C 6 heterocycloalkenyl, and C 5 -C 6 heterocycloalkynyl.
  • Aryl molecules include, for example, cyclic hydrocarbons that incorporate one or more planar sets of, typically, six carbon atoms that are connected by delocalized electrons numbering the same as if they consisted of alternating single and double covalent bonds.
  • An example of an aryl molecule is benzene.
  • Heteroaryl molecules include substitutions along their main cyclic chain of atoms such as O, N, or S. When heteroatoms are introduced, a set of five atoms, e.g., four carbon and a heteroatom, can create an aromatic system.
  • heteroaryl molecules include furan, pyrrole, thiophene, imadazole, oxazole, pyridine, pyrazole, and pyrazine.
  • Aryl and heteroaryl molecules can also include additional fused rings, for example, benzofuran, indole, benzothiophene, naphthalene, anthracene, and quinoline.
  • alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl molecules used herein can be substituted or unsubstituted.
  • the term substituted includes the addition of an alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl group to a position attached to the main chain of the alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl, e.g., the replacement of a hydrogen by one of these molecules.
  • substitution groups include, but are not limited to, hydroxyl, halogen (e.g., F, Br, Cl, or I), and carboxyl groups.
  • halogen e.g., F, Br, Cl, or I
  • carboxyl groups e.g., but are not limited to, hydroxyl, halogen (e.g., F, Br, Cl, or I), and carboxyl groups.
  • the term unsubstituted indicates the alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl has a full complement of hydrogens, i.e., commensurate with its saturation level, with no substitutions, e.g., linear decane (—(CH 2 ) 9 —CH
  • the compounds described herein can be prepared in a variety of ways known to one skilled in the art of organic synthesis or variations thereon as appreciated by those skilled in the art.
  • the compounds described herein can be prepared from readily available starting materials. Optimum reaction conditions may vary with the particular reactants or solvents used, but such conditions can be determined by one skilled in the art.
  • Variations on the Formula I include the addition, subtraction, or movement of the various constituents as described for each compound. Similarly, when one or more chiral centers are present in a molecule, the chirality of the molecule can be changed.
  • the compounds described herein can be isolated in pure form or as a mixture of isomers. Additionally, compound synthesis can involve the protection and deprotection of various chemical groups. The use of protection and deprotection, and the selection of appropriate protecting groups can be determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4th Ed., Wiley & Sons, 2006, which is incorporated herein by reference in its entirety.
  • Reactions to produce the compounds described herein can be carried out in solvents, which can be selected by one of skill in the art of organic synthesis.
  • Solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products under the conditions at which the reactions are carried out, i.e., temperature and pressure. Reactions can be carried out in one solvent or a mixture of more than one solvent.
  • Product or intermediate formation can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry
  • HPLC high performance liquid chromatography
  • kits for treating or preventing a sexually transmitted infection in a subject comprise administering to the subject the compounds described herein, wherein the compounds bind the SEVI-fibrils to inhibit the ability of the SEVI-fibrils to enhance a sexually transmitted infection.
  • Compounds contained within International Publication No. WO 2007/011834 are also contemplated herein for use in methods of treating or preventing a sexually transmitted infection.
  • the methods optionally, comprise identifying a subject with or at risk of developing a sexually transmitted infection and administering to the subject a semen-derived enhancer of viral infection (SEVI)-binding small molecule.
  • SEVI semen-derived enhancer of viral infection
  • the methods can further comprise administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
  • the SEVI-binding small molecule can, for example, comprise a hydrophobic molecule, wherein the hydrophobic molecule incorporates into and binds the SEVI-fibrils.
  • SEVI-fibrils are formed as a result of ahydrophobic interactions between component monomer polypeptides.
  • exogenous hydrophobic molecules such as hydrophobic polypeptides
  • hydrophobic molecules include alkanes, oils, fats, and greasy substances in general.
  • the SEVI-binding small molecule can, for example, comprise an anionic polypeptide supramolecular assembly.
  • the anionic supramolecular assembly is water-soluble.
  • the anionic supramolecular assembly comprises a soluble hydrogel and other supramolecular assemblies derived from an Ac-(XEXE)n-NH2 (SEQ ID NO:14) polypeptide and related polypeptides.
  • Water-soluble supramolecular assemblies derived from self-assembling anionic polypeptides can, for example, bind to the cationic SEVI fibrils and inhibit interactions between the SEVI-fibrils and the infectious agents.
  • An example of a soluble hydrogel is the PuraMatrix hydrogel.
  • the PuraMatrix hydrogel comprises a (VKVK)n polypeptide fibrillar hydrogel that is not toxic.
  • the SEVI-binding small molecules can further comprise a bulky side chain, a negatively charged side chain, a coupled moiety, and an anti-viral molecule.
  • a bulky side chain can, for example, comprise a poly-ethylene glycol (PEG) molecule.
  • An anti-viral molecule can, for example, comprise a pradimicin A or AZT molecule.
  • Methods of screening for agents that are capable of binding SEVI-fibrils include the steps of providing the agent to be screened, contacting the agent with the SEVI-fibrils, and determining whether the agent to be screened binds the SEVI-fibrils. Binding can be determined, for example, by selecting an assay from the group consisting of a coimmunoprecipitation assay, a colocalization assay, or a fluorescence polarizing assay, as described below.
  • the assays are known in the art, e.g., see Sambrook et al., Molecular Cloning: A Laboratory Manual, 3 rd Ed., Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (2001); Dickson, Methods Mol. Biol. 461:735-44 (2008); Nickels, Methods 47(1):53-62 (2009); and Zinchuk et al., Acta Histochem. Cytochem. 40(4):101-11 (2007).
  • the SEVI-binding agents, SEVI-binding small molecules, anti-viral agents, anti-bacterial agents, anti-fungal agents described herein or derivatives thereof can be provided in a pharmaceutical composition.
  • the pharmaceutical composition can be in the form of solid, semi-solid or liquid dosage forms, such as, for example, tablets, suppositories, pills, capsules, powders, liquids, or suspensions, preferably in unit dosage form suitable for single administration of a precise dosage.
  • the compositions will include a therapeutically effective amount of the compound described herein or derivatives thereof in combination with a pharmaceutically acceptable carrier and, in addition, may include other medicinal agents, pharmaceutical agents, carriers, or diluents.
  • pharmaceutically acceptable is meant a material that is not biologically or otherwise undesirable, which can be administered to an individual along with the selected compound without causing unacceptable biological effects or interacting in a deleterious manner with the other components of the pharmaceutical composition in which it is contained.
  • compositions are optionally, provided in the form of contraceptives or contraceptive agents, such as condoms or spermicides, or lubricants.
  • the term carrier encompasses any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, lipid, stabilizer, or other material well known in the art for use in pharmaceutical formulations.
  • a carrier for use in a composition will depend upon the intended route of administration for the composition.
  • the preparation of pharmaceutically acceptable carriers and formulations containing these materials is described in, e.g., Remington's Pharmaceutical Sciences, 21st Edition, ed. University of the Sciences in Philadelphia, Lippincott, Williams & Wilkins, Philadelphia Pa., 2005.
  • physiologically acceptable carriers include buffers such as phosphate buffers, citrate buffer, and buffers with other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN® (ICI, Inc.; Bridgewater, N.J.), polyethylene glycol (PEG), and PLURONICSTM (BASF; Florham Park, N.J.).
  • buffers such as phosphate buffers, citrate buffer, and buffers with
  • compositions containing the compound described herein or derivatives thereof suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispensing agents.
  • adjuvants such as preserving, wetting, emulsifying, and dispensing agents.
  • Prevention of the action of microorganisms can be promoted by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • Isotonic agents for example, sugars, sodium chloride, and the like may also be included.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Solid dosage forms for oral administration of the compounds described herein or derivatives thereof include capsules, tablets, pills, powders, and granules.
  • the compounds described herein or derivatives thereof is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid
  • binders as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose, and acacia
  • humectants as for example, glycerol
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate
  • solution retarders as for example, paraffin
  • absorption accelerators as for example, paraffin
  • compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethyleneglycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others known in the art. They may contain opacifying agents and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes. The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration of the compounds described herein or derivatives thereof include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents, and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols, and fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • inert diluents commonly used in the art
  • composition can also include additional agents, such as wetting, emulsifying, suspending, sweetening, flavoring, or perfuming agents.
  • additional agents such as wetting, emulsifying, suspending, sweetening, flavoring, or perfuming agents.
  • Suspensions in addition to the active compounds, may contain additional agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • additional agents as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • compositions of the compounds described herein or derivatives thereof for rectal administrations are preferably suppositories, which can be prepared by mixing the compounds with suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
  • Dosage forms for topical administration of the compounds described herein or derivatives thereof include ointments, powders, sprays, gels and the like.
  • the compounds described herein or derivatives thereof are admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as may be required.
  • salts refers to those salts of the compound described herein or derivatives thereof that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds described herein.
  • salts refers to the relatively non-toxic, inorganic and organic acid addition salts of the compounds described herein. These salts can be prepared in situ during the isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate, methane sulphonate, and laurylsulphonate salts, and the like.
  • alkali and alkaline earth metals such as sodium, lithium, potassium, calcium, magnesium, and the like
  • non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • the active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount.
  • Those of skill in the art will understand that the specific dose level and frequency of dosage for any particular subject may be varied, and it will be understood that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and response of the individual subject, the severity of the subject's symptoms, and the like.
  • compositions described herein are useful for preventing or reducing the transmission of sexually transmitted infections (STIs).
  • STIs sexually transmitted infections
  • administering a SEVI-binding agent or SEVI-binding small molecule to a subject interferes with the binding of infectious agents to the semen fibrils. Such binding interferes with the infection-enhancing activity of the semen fibrils and prevents or reduces the risk of STIs.
  • the compositions are useful for treatment prior to, during, or after infection. Treatment can completely or partially abolish some or all of the signs and symptoms of transmission of the infection and reduce the likelihood that the treated subject will subsequently develop symptoms of an STI or will delay the onset of symptoms. Thus, for example, treatment can prevent, reduce or delay viral transmission, e.g., HIV transmission.
  • STIs are infections that can be transferred from one subject to another through sexual contact.
  • STIs are caused by microorganisms that are transmitted via semen, vaginal secretions or blood during sexual contact or by microorganisms that survive on the skin and mucous membranes of the genital area.
  • sexual contact can include sexual intercourse (vaginal and anal), oral-genital contact, and the use of sexual toys, such as vibrators.
  • Microorganisms transmitted via sexual contact can include, for example, viruses, e.g., HIV, human papilloma virus (HPV), herpesviruses, hepatitis B, and C and cytomegalovirus (CMV); bacteria, e.g., infectious agents responsible for gonorrhea ( Neisseria gonorrhoeae ); syphilis ( Treponema pallidum ); chancroid ( Haemophilus ducreyi ); donovanosis ( Granuloma inguinale or Calymmatobacterium granulomatis ); lymphogranuloma venereum (LGV) ( Chlamydia trachomatis ); non-gonococcal urethritis (NGU) ( Ureaplasma urealyticum or Mycoplasma hominis ); bacterial vaginosis and Staphylococcus aureus ; protozoa, e.g., infectious
  • STIs Symptoms of STIs can vary and often the infected subject has no symptoms. However, an asymptomatic subject may be able to pass the disease to a sexual partner. Common symptoms of STI's include, but are not limited to, urethral discharge, genital ulcers, inguinal swellings, scrotal swelling, vaginal discharge, lower abdominal pain, fever, lymphadenopathy (swollen lymph nodes), pharyngitis (sore throat), rash, myalgia (muscle pain), malaise, and mouth and esophageal sores. Both symptomatic and asymptomatic infections can lead to the development of more serious conditions, including AIDS, pelvic inflammatory disease, infertility and tubal (ectopic) pregnancy, genital warts, cervical and other genital cancers.
  • compositions and methods are applicable to the transmission of infections by any type of HIV, e.g., HIV-1 and HIV-2.
  • the compositions can be administered to both men and women.
  • the compositions are suitable for a subject who is not infected with HIV, but is at risk for sexually transmitted infection.
  • Subjects who may be at increased risk of becoming infected through sexual contact include those who have unprotected sex, i.e., do not use condoms during sexual intercourse; have multiple sex partners; males who have sexual intercourse with other men; those who have high-risk partner(s), i.e., the sexual partner has multiple sex partners, is a man who has sex with other men, or is an intravenous drug user; or those who have or have recently had a sexually transmitted disease, e.g., syphilis, gonorrhea of chlamydia.
  • a sexually transmitted disease e.g., syphilis, gonorrhea of chlamydia.
  • compositions are also useful in an infected subject, e.g., a subject who has an HIV infection, to reduce the transmission to an uninfected partner.
  • the compositions can be administered to a subject at any stage in the course of HIV infection.
  • compositions can be monitored according to standard methods in the art for assessing HIV status, including measuring the level of HIV, using for example a PCR assay, in a clinical sample, e.g., a blood sample, measuring the level of anti-HIV antibodies, using for example, an ELISA or immunoblotting assay, in a clinical sample, e.g., a blood sample, and by monitoring the levels of CD4+ T cells in a clinical sample.
  • compositions can be administered in conjunction with other therapeutic or prophylactic modalities to an individual in need of treatment.
  • the compositions may be administered to a subject who practices “safe sex”, i.e., a subject who wears a condom during sexual intercourse or has sexual intercourse with a partner who wears a condom.
  • safe sex i.e., a subject who wears a condom during sexual intercourse or has sexual intercourse with a partner who wears a condom.
  • the condom can be disguised to contain or be coated with the therapeutic agent.
  • compositions can also be administered in conjunction with other therapies for treating HIV infection, such as standard small molecule pharmaceutical agents, topical microbicides, biopharmaceuticals (e.g., antibodies or antibody-related immunotherapies, siRNAs, shRNAs, antisense oligonucleotides and other RNA inhibitory molecules, microRNAs, and peptide therapeutics), surgery, or in conjunction with any medical devices that may be used to assist the subject.
  • standard therapy for HIV infection includes highly active antiretroviral therapy, or HAART.
  • HAART includes a combination (or “cocktail”) of drugs belonging to at least two classes of antiretroviral agents, e.g., a nucleoside analogue reverse transcriptase inhibitors (NARTIs or NRTIs), a non-nucleoside reverse transcriptase inhibitor and a protease inhibitor.
  • NARTIs or NRTIs nucleoside analogue reverse transcriptase inhibitors
  • NRTIs Non-nucleoside reverse transcriptase inhibitors
  • NTP Nevirapine
  • DLV Delavirdine
  • Efavirenz Efavirenz
  • ETV Intelence
  • Protease inhibitors include, for example: Saquinavir (SQV, Invirase), Indinavir (IDV, Crixivan), Ritonavir (RTV, Norvir), Nelfinavir (NFV, Viracept), Amprenavir (APV, Agenerase), Lopinavir (LPV, Kaletra, Aluvia), Atazanavir (ATV, Reyataz), Fosamprenavir (FPV, Lexiva), Tipranavir (TPV, Aptivus) and Darunavir (DRV, Prezista).
  • anti-HIV drugs include fusion and attachment inhibitors, including, for example, Enfuvirtide (Fuzeon or T-20) and Maraviroc (MVC, Selzentry, Celsentri); and integrase inhibitor, including for example, Raltegravir (RGV, Isentress).
  • fusion and attachment inhibitors including, for example, Enfuvirtide (Fuzeon or T-20) and Maraviroc (MVC, Selzentry, Celsentri); and integrase inhibitor, including for example, Raltegravir (RGV, Isentress).
  • the compositions can be incorporated into standard barrier prophylatics, for example male and female condoms.
  • the duration of treatment with any composition provided herein can be any length of time from as short as one day to as long as the life span of the host (e.g., many years).
  • the composition can be administered once a week (for, for example, 4 weeks to many months or years); once a month (for, for example, three to twelve months or for many years); or once a year for a period of 5 years, ten years, or longer.
  • the frequency of treatment can be variable.
  • the compositions can be administered once (or twice, three times, etc.) daily, weekly, monthly, or yearly.
  • an effective amount of any composition provided herein can be administered to an individual in need of treatment.
  • the term “effective” as used herein refers to any amount that induces a desired response while not inducing significant toxicity in the patient. Such an amount can be determined by assessing a patient's response after administration of a known amount of a particular composition.
  • the level of toxicity if any, can be determined by assessing a patient's clinical symptoms before and after administering a known amount of a particular composition. It is noted that the effective amount of a particular composition administered to a patient can be adjusted according to a desired outcome as well as the patient's response and level of toxicity. Significant toxicity can vary for each particular patient and depends on multiple factors including, without limitation, the patient's disease state, age, and tolerance to side effects.
  • clinical methods that can assess the degree of a particular disease state can be used to determine if a response is induced.
  • blood or laboratory tests may be administered to determine HIV titers before, during and after a course of treatment.
  • the particular methods used to evaluate a response will depend upon the nature of the patient's disorder, the patient's age, and sex, other drugs being administered, and the judgment of the attending clinician.
  • the semen fibrils comprise fibrillary aggregates derived from polypeptides in seminal fluid.
  • the fibrillary aggregates can be insoluble fibrous protein aggregates that are generally characterized by a cross-beta sheet quaternary structure; i.e., a monomeric unit contributes a beta strand to a beta sheet, which spans across more than one molecule.
  • the fibrils can be identified using a variety of assays, including fluorescent dyes, e.g., thioflavin T binding, Congo red staining, stain polarimetry, circular dichroism, FTIR or X-ray diffraction analysis.
  • X-ray diffraction analysis reveals characteristic scattering diffraction signals produced at 4.7 and 10.6 ⁇ ngstroms (0.47 nm and 1.06 nm), corresponding to the interstrand and stacking distances in beta sheets.
  • the stacks of beta sheet are short and traverse the breadth of the amyloid fibril; the length of the fibril is built by aligned strands.
  • Semen fibrils can form from semen fibrillary polypeptides or oligomers thereof.
  • a semen fibrillary polypeptide can be a fibril forming fragment of prostatic acid phosphatase (PAP), a protein produced by the prostate and secreted into semen.
  • PAP also known as ACP-3 or prostatic acid phosphatase precursor 3, ACP3, ACPP or EC 3.1.3.2
  • ACP3 prostatic acid phosphatase precursor 3
  • ACPP prostatic acid phosphatase precursor 3
  • EC 3.1.3.2 is the prostate-specific form of one of five ubiquitous acid phosphatase isozymes that catalyze the conversion of orthophosphoric monoester to alcohol and orthophosphate.
  • PAP is over 100 times more abundant in the prostate that in other tissue types.
  • subject can be a vertebrate, more specifically a mammal (e.g. a human, horse, cat, dog, cow, pig, sheep, goat, mouse, rabbit, rat, and guinea pig), birds, reptiles, amphibians, fish, and any other animal.
  • a mammal e.g. a human, horse, cat, dog, cow, pig, sheep, goat, mouse, rabbit, rat, and guinea pig
  • the term does not denote a particular age or sex. Thus, adult and newborn subjects, whether male or female, are intended to be covered.
  • patient or subject may be used interchangeably and can refer to a subject with a sexually transmitted infection.
  • patient or subject includes human and veterinary subjects.
  • treatment refers to a method of reducing the effects of a sexually transmitted infection or a symptom of the sexually transmitted infection as described above.
  • treatment can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% reduction in the severity of a sexually transmitted infection or a symptom of the sexually transmitted infection.
  • a method for treating a sexually transmitted infection is considered to be a treatment if there is a 10% reduction in one or more symptoms of the infection in a subject as compared to a control.
  • the reduction can be a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or any percent reduction in between 10% and 100% as compared to native or control levels. It is understood that treatment does not necessarily refer to a cure or complete ablation of the infection or symptoms of the infection.
  • the primary human macrophages were stimulated with either LPS (100 ng/mL), SEVI (10 mM), or both.
  • Cell culture supernatants were collected at 0, 4, and 24-hour timepoints and measurements of TNF ⁇ and IL-1 ⁇ were determined by ELISA. Briefly, 96-well plates were coated with 100 ⁇ L/well of capture antibody in coating buffer (eBioscience, Inc.; San Diego, Calif.) and incubated overnight at 4° C. The wells were washed with phosphate buffered saline (PBS) with 0.05% Tween-20 and blocked for 1 hour with 300 ⁇ L/well assay diluent (eBioscience, Inc.).
  • PBS phosphate buffered saline
  • Optical density was read at 450 nm with a SpectraCount plate reader (Packard Instrument Company; Meriden, Conn.), and the cytokine levels were then calculated by extrapolation to a standard curve generated using known amounts of recombinant IL-1 ⁇ and TNF ⁇ .
  • BTA-EG 6 was synthesized as described previously (Inbar, P., Li, C. Q., Takayama, S. A., Bautista, M. R., and Yang, J. (2006) Chembiochem 7, 1563-1566).
  • CEM-M7 (a gift from N. Landau, New York University, New York, N.Y.) and Jurkat cells were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum, penicillin (50 units/ml), and streptomycin (50 ⁇ g/ml).
  • SiHa cells were cultured in DMEM medium supplemented with 10% fetal bovine serum, penicillin (50 units/ml), and streptomycin (50 ⁇ g/ml).
  • A2En cells (a gift from S. Greene, Louisiana State University Health Sciences Center, New Orleans, La.), and 3EC1 cells (a gift from R.
  • PBMCs were isolated from whole blood by Lymphoprep density gradient centrifugation. PBMCs were stimulated for 48 h in RPMI 1640 medium supplemented with 5% human IL-2 (ZeptoMetrix, Buffalo, N.Y.), 5 ⁇ g/ml PHA (Sigma, St. Louis, Mo.), 20% fetal bovine serum, penicillin (50 units/ml), and streptomycin (50 ⁇ g/ml).
  • PAP248-286 and biotinylated PAP248-286 in which a biotin was added to the amino terminus of the peptide, was synthesized and dissolved in PBS at a concentration of 10 mg/ml. Fibrils were formed by agitation in an Eppendorf Thermomixer at 1400 rpm (Eppendorf, Hauppauge, N.Y.) and 37° C. for 72 h. Semen samples were obtained from the Strong Fertility Center (Rochester, N.Y.) and Fairfax CryoBank (Fairfax, Va.). Samples were pooled, aliquoted, and stored at ⁇ 80° C.
  • Binding of BTA-EG 6 to SEVI fibrils was measured according to the centrifugation assay described by Levine (LeVine, H., 3rd (2005) Amyloid 12: 5-14) for BTA-1 to A ⁇ fibrils. Briefly, 200 ⁇ l of various concentrations of BTA-EG 6 in PBS were incubated in the presence or absence of 10 ⁇ g of SEVI fibrils to give a final volume of 220 ⁇ l of solution. These incubations were performed in duplicate runs and allowed to equilibrate overnight at room temperature. After equilibration, each solution was centrifuged at 16,000 ⁇ g for 30 min.
  • the data were processed using Origin 7.0 (MicroCal Software, Inc., Northampton, Mass.).
  • Jurkat cells were incubated with biotinylated SEVI fibrils (40 ⁇ g/ml) with and without BTA-EG6 at a concentration of 10 (low) or 30 ⁇ g/ml (high) or heparin (100 ⁇ g/ml) as a positive control for interfering with SEVI binding to the cell surface.
  • Cells were incubated for 1 h at 37° C., washed, and stained for 1 h with a covalent conjugate of streptavidin and fluorescein isothiocyanate (SA-FITC). Cells were washed and run on an Accuri C6 Flow Cytometer (Accuri Cytometers, Ann Arbor, Mich.). Data were analyzed using FlowJo (TreeStar Inc, Ashland, Oreg.).
  • X4 tropic HIV-1 IIIB 21 ng/ml p24
  • R5 tropic HIV-1 ADA 60 ng/ml p24
  • Treated virions were then added to 5 ⁇ 10 4 CEM-M7 cells/well in 96-well flat-bottomed tissue culture plates. After 2 h, the medium axis were replaced. Infection was assayed after 48 h by quantifying luciferase expression using the Promega Dual-Luciferase assay and a Beckman Coulter DTX880 plate reader.
  • pooled human semen samples were added to virions at a 1:1 dilution and incubated for 15 min at room temperature in the presence or absence of BTA-EG 6 . After 15 min, the semen and virus mixture was diluted 1:15 into 5 ⁇ 10 4 CEM-M7 cells/well in a 96-well plate. Cells were washed after 1 h, and infection was assayed at 48 h as above.
  • R5 tropic HIV-1BaL preincubated with 15 ⁇ g/ml SEVI in the presence or absence of BTA-EG6 was added to 2 ⁇ 105 PHA/IL-2-stimulated PBMCs/well in 96-well flat-bottomed tissue culture plates. Cells were washed at 3 h, and infection was analyzed at day 4 using the HIV-1 p24 antigen capture assay (Advanced Bioscience Laboratory).
  • HIV-1 IIIB or ADA virions were pretreated with 15 ⁇ g/ml SEVI and added to 5 ⁇ 10 4 Jurkat cells, or A2En cells, in the presence or absence of BTA-EG6. After 90 min, cells were washed to remove any unbound virus, and bound virions were detected using an HIV-1 p24 antigen capture assay (Advanced Bioscience Laboratory).
  • HIV-1 BaL virions were pretreated with 15 ⁇ g/ml SEVI and added to 5 ⁇ 10 4 A2En cells in the presence or absence of BTA-EG6. Supernatant was collected at 6 and 24 h, and the production of the chemokines IL-8 and Mip-3 ⁇ was measured by ELISA (R&D Systems). To assess semen-mediated chemokine production, SiHa cells were treated with semen, as described above, in the presence or absence of BTA-EG6. After 6 h, supernatants were collected, and the production of the chemokines IL-8 and Mip-3 ⁇ was measured by ELISA (R&D systems, Minneapolis, Minn.).
  • the cervical epithelial cell lines SiHa, A2En (endocervical), and 3EC1 (ectocervical) were treated for 12 h with BTA-EG 6 at concentrations up to 66 ⁇ g/ml, 10 times the IC 50 .
  • cell viability was analyzed by measuring cellular metabolic activity using the resazurin cytotoxicity assay, AlamarBlue® (Invitrogen), in accordance with the manufacturer's protocol. Cytokine and chemokine production was assessed at 12 h by ELISA (R&D systems).
  • Cells were also treated with 0.1% nonoxynol-9 as a positive control for cytotoxicity and with 0.1 ⁇ g/ml FSL1, a synthetic diacylated lipoprotein derived from Mycoplasma salivarium (InvivoGen, San Diego, Calif.), as a positive control for chemokine production.
  • FSL1 a synthetic diacylated lipoprotein derived from Mycoplasma salivarium
  • SEVI fibrils are highly cationic, with a negative charge of +6.5 at neutral pH and +8 at pH 5, as would be seen in the vaginal mucosa.
  • the cationic nature of SEVI is required for its ability to enhance HIV infection. This suggests that SEVI acts in a manner similar to other cationic polymers to enhance HIV infectivity.
  • HIV-1 IIIB virions were preincubated with 50% semen and increasing concentrations of BTA-EG 4 and BTA-EG 6 . After 10 minutes, the stocks were diluted 15 fold and incubated with CEM 5.25 cells. The increasing concentrations of BTA-EG 4 and BTA-EG 6 resulted in a decrease in luciferase activity ( FIGS. 6A and 6B ), indicating that BTA-EG 4 and BTA-EG 6 were capable of inhibiting semen mediated enhancement of HIV infection. It was further found that BTA-EG 4 and BTA-EG 6 were capable of inhibiting SEVI-enhanced binding of HIV to the cell surface.
  • HIV-1 IIIB virions were pretreated with 10 ⁇ g/mL SEVI and added to Jurkat cells with or without increasing concentrations of BTA-EG 4 and BTA-EG 6 . After 90 minutes cells were washed to remove any unbound virus and bound virions were detected using a p24 ELISA. Increasing concentrations of BTA-EG 4 and BTA-EG 6 resulted in a decrease in HIV-1 binding to the cells ( FIGS. 7A and 7B ).
  • a fluorescence polarizing screen was developed. The screen is shown in FIG. 8 . Polarized light passed over a small unbound molecule with a fluorescent moiety will produce rapid rotation and will result in fluorescence that is de-polarized. Polarized light passed over SEVI bound to a small molecule with a fluorescent moiety will result in fluorescence that is polarized.
  • BTA-EG 4 and BTA-EG 6 were capable of being administered to cervical epithelial cells without cytotoxicity
  • the cervical cell lines A2En (endocervical) and SiHa cells were treated with BTA-EG 4 and BTA-EG 6 for 12 hours at concentrations up to 10 times greater than the inhibitory concentration.
  • viability was measured with Alamar Blue and it was shown that BTA-EG 4 and BTA-EG 6 did not affect the cell viability of A2En and SiHa cells. It was further shown in SiHa cells that BTA-EG 4 and BTA-EG 6 do not induce cytokine production.
  • SiHa cells were treated with BTA-EG 4 and BTA-EG 6 at varying concentrations for 6 hours and cytokine production was examined by ELISA. As shown in FIGS. 11A-11C , IL-1b, MIP-3a, and TNF-a levels were unaffected by varying concentrations of BTA-EG 4 and BTA-EG 6 .
  • ThT is able to intercalate into the generic ⁇ -sheet structure of amyloid fibrils.
  • the benzothiazole aniline derivative, BTA-EG 6 is a ThT analog carrying a hexa(ethylene glycol) moiety ( FIG. 12A ). This molecule binds to A ⁇ fibrils and interferes with the ability of A ⁇ -binding proteins to interact with the fibrils. Fluorescence polarization was used to measure the ability of BTA-EG 6 to bind SEVI. Increasing concentrations of BTA-EG 6 were added to 50 ⁇ g/ml SEVI that had been preincubated with 16 ⁇ g/ml FITC-heparin, a known SEVI binder. BTA-EG 6 was able to displace fluorescent heparin from the SEVI fibrils in a dose-dependent fashion ( FIG. 12B ), thus showing an interaction between these molecules and the fibrils.
  • ThT fluorescence changes in fluorescence intensity when intercalated into the ⁇ -sheet structure common to amyloid fibrils; therefore, ThT fluorescence serves as a surrogate measure for fibrillar structure of SEVI and for the stability of SEVI fibrils.
  • FIG. 12D the addition of BTA-EG 6 had no effect on fibrillar stability as measured by ThT. Unlike in the case with ThT, the fluorescence intensity of BTA-EG 6 does not change upon binding to amyloid fibrils.
  • Binding of the fibrils to the cell surface was detected using SA-FITC.
  • FIG. 12E and Table 1 increasing concentrations of BTA-EG 6 inhibited the ability of SEVI fibrils to interact with and bind the cell surface, as measured by both the percentage of cells with bound fibrils and the mean fluorescence intensity of the cells.
  • Table 1 shows that BTA-EG 6 binding to SEVI inhibits interaction of SEVI fibrils with the cell surface.
  • Jurkat cells were incubated with SEVI-biotin (SEVI-Bio) for 1 h in the presence or absence of 5.5 (low) or 27 ⁇ g/ml (high) BTA-EG 6 .
  • Surface-bound fibrils were detected with SA-FITC and measured by flow cytometry. Results are shown as percentage of cells with bound fibrils (SA-FITC+) as well as mean fluorescent intensity (MFI).
  • CEM-M7 cells were infected with HIV-1 strain IIIB plus 15 ⁇ g/ml SEVI fibrils in the presence of increasing concentrations of BTA-EG 6 .
  • CEM-M7 cells are a CD4 + CCR5 + CXCR4 + T/B cell hybrid cell line and contain HIV LTR-driven luciferase and GFP reporter gene cassettes.
  • the HIV LTR is a weak transcriptional regulator in the absence of its cognate, virally encoded trans-activator, Tat.
  • BTA-EG 6 was able to effectively inhibit SEVI-mediated enhancement of HIV infection in a dose-dependent fashion, reducing infectivity nearly back to baseline levels (i.e. levels detected in the absence of SEVI) at the highest concentrations tested ( FIG. 13A ). Importantly, BTA-EG 6 had no effect on the infectivity of HIV virus alone, even at the highest concentrations ( FIG. 13B ), indicating that this effect was not due to direct inhibition of intrinsic virus infectivity.
  • CEM-M7 cells were infected with HIV-1ADA and 15 ⁇ g/ml SEVI in the presence of BTA-EG 6 .
  • Ten different BTA-EG 6 concentrations were tested, ranging from 0.4 to 50 ⁇ g/ml.
  • the data were fit to an exponential decay curve to calculate the IC 50 , and results are shown in FIG. 13D .
  • the calculated IC 50 was 6.6 ⁇ g/ml for BTA-EG 6 (equivalent to 13 ⁇ M).
  • BTA-EG 6 For BTA-EG 6 to be a viable microbicide candidate, it must be effective not just against the effects of SEVI but should be able to effectively inhibit the infection-enhancing activity of human semen. Therefore, the effect of this compound on semen-mediated enhancement of HIV-1 infection was examined. As human semen has been reported to be toxic to cultured cells, a protocol that minimized this toxicity was used. Pooled human semen was added to HIV-1IIIB virus in a 1:1 dilution. After 15 min, this solution was added to cells at a 1:15 dilution for a final concentration of 3.3%. As shown in FIG. 14A , BTA-EG 6 efficiently inhibited the semen mediated enhancement of HIV-1 infection at similar concentrations to those active against SEVI alone. FIG. 14B shows that this effect extended to infection with an R5 virus, HIV ADA as well.
  • BTA-EG 6 inhibited semen-mediated chemokine release was examined.
  • Human semen can be pro-inflammatory, mediating the release of IL-8 and MIP-3 ⁇ from cervical endothelial cells. This property is thought to be due to the presence of several pro-inflammatory mediators but is not due to the presence of SEVI as SEVI does not stimulate the release of IL-8 or MIP-3 ⁇ .
  • BTA-EG 6 Inhibits SEVI-Mediated Attachment of HIV-1 to the Cell Surface
  • SEVI was able to strongly enhance the binding of virions to the cell surface, and this effect was efficiently abrogated by BTAEG 6 ( FIG. 15A ).
  • BTA-EG 6 had no effect on the binding of HIV virions to the cell surface in the absence of SEVI ( FIG. 15A ).
  • Similar results were obtained with an R5 virus, HIV-1ADA ( FIG. 4B ).
  • this experiment was performed using A2En cells, a primary cell-derived endocervical cell line. It was found that SEVI also enhanced binding of virions to the surface of these cervical epithelial cells and that this effect was inhibited by BTA-EG 6 ( FIG. 15C ).
  • BTA-EG 6 is not Toxic to Cervical Cells
  • FIG. 16A shows that BTA-EG 6 did not have any effects on cell viability, even at the highest concentrations tested.
  • Nonoxynol-9 (non-9), a spermicide, was used as a positive control. Whether treatment with BTA-EG 6 led to the production of inflammatory cytokines and chemokines from the cervical cell lines was examined. All three cervical cell lines were treated for 6 h with concentrations of BTA-EG 6 ranging from 6.6 to 66 ⁇ g/ml. Cell culture supernatants were then assessed for the presence of the inflammatory cytokines and chemokines Mip-3 ⁇ ( FIG. 5B ), IL-8 ( FIG. 16C ), IL-1 ⁇ , and TNF- ⁇ .
  • cytokines and chemokines were selected because they are up-regulated by other candidate microbicides and because they may play a role in microbicide-mediated enhancement of HIV-1 infection.
  • BTA-EG 6 did not lead to the release of any of these cytokines or chemokines, even at the highest doses tested. These results indicate that BTA-EG 6 is not toxic to cervical endothelial cells.
  • BTA-EG 6 inhibited SEVI-mediated enhancement of infection by both X4 (HIV-1 IIIB ) and R5 (HIV-1 ADA ) strains, in a dose-dependent fashion.
  • HIV-1 ADA the IC50 was 13 ⁇ M; this value is 100-fold higher than the measured Kd of BTA-EG 6 for binding to aggregated SEVI peptides (127 nM).
  • one explanation for this difference is that the ability of BTA-EG 6 to compete with virion/fibril or virion/cell interactions requires a greater number of BTA-EG 6 molecules than the noncompetitive binding of BTA-EG 6 to SEVI alone.
  • BTA-EG 6 also inhibited SEVI-enhanced infection of primary cells (human peripheral blood mononuclear cells) in a dose-dependent fashion, and it blocked SEVI-enhanced binding of X4 (HIV-1 IIIB ) and R5 (HIV-1 ADA ) strains to target cells (including both Jurkat T cells and A2En endocervical cells).
  • primary cells human peripheral blood mononuclear cells
  • R5 HIV-1 ADA
  • BTA-EG 6 and related compounds not only reduce the efficiency of HIV-1 infection of target cells but also reduce the level of target cell recruitment to virus-exposed genital mucosal tissue.
  • BTA-EG6 effectively prevents semen mediated enhancement of HIV infectivity, showing that this activity of semen can be targeted by specifically inhibiting the SEVI fibrils.
  • BTA-EG6 did not inhibit other properties of semen, such as the ability to elicit pro-inflammatory chemokines
  • BTA-EG6 is an effective microbicide target
  • HIV-1 IIIB virions were pretreated with 15 ⁇ g/ml SEVI and added to 5 ⁇ 10 4 A2En cells (immortalized primary human endocervical cells) ( FIG. 17A ) or to Jurkat T cells (a CD4+ human T cell line) ( FIG. 17B ) in the presence or absence of test compound BTA-EG 6 in monomeric, dimeric, trimeric, tetrameric or pentameric forms (at a final concentration of 25 ⁇ M). After 90 min, cells were washed to remove any unbound virus, and bound virions were detected using an HIV-1 p24 antigen capture assay (Advanced Bioscience Laboratory, Rockville, Md.). The data showed reduced HIV-1 p24 antigen capture in the presence of SEVI as compared to capture in the absence of SEVI.
  • FIG. 18 shows the structure of a benzothiazole aniline (BTA)-based monomer (1), dimer (2), trimer (3), tetramer (4), and pentamer (5) and a schematic of how the scaffold may affect binding.
  • BTA benzothiazole aniline

Abstract

Described herein are compositions and methods for treating or preventing a sexually transmitted infection in a subject.

Description

    STATEMENT REGARDING FEDERALLY FUNDED RESEARCH
  • This invention was made with government support under Grant Nos. R21 AI074351, RO1AI084111, and T32 DA07232 from the National Institutes of Health. The United States government has certain rights in this invention.
  • BACKGROUND
  • There are over twenty types of sexually transmitted infections (STI) associated with various bacteria, protozoa, fungi and viruses. One example of a viral sexually transmitted infection is human immunodeficiency virus (HIV). Acquired immunodeficiency syndrome (AIDS) is a collection of symptoms and infections resulting from the specific damage to the immune system caused by HIV. In 2006, nearly 25 years after the first report of AIDS cases, there were over 39 million persons living with HIV infection worldwide. About one-fourth of those with infected with HIV have not yet been diagnosed and are unaware of their status. AIDS has become one of the deadliest epidemics in human history, killing more than 25 million people around the world. In the last decade, major advances in prevention and treatment for HIV/AIDS have prolonged and improved the lives of many, but despite such advances, an estimated 4 million people still become infected with HIV every year, and many of these are people under the age of 25. In 2006, HIV/AIDS was responsible for nearly 3 million deaths worldwide.
  • SUMMARY
  • Provided herein are methods of treating or preventing a sexually transmitted infection in a subject. The methods comprise administering to a subject with or at risk of acquiring a sexually transmitted infection a semen-derived enhancer of viral infection (SEVI)-binding agent comprising a compound described herein, including, e.g., BTA-EG4 and BTA-EG6. Also provided are methods comprising administering to a subject with or at risk of acquiring a sexually transmitted infection a semen-derived enhancer of viral infection (SEVI)-binding small molecule. The SEVI-binding small molecule can, for example, comprise a hydrophobic molecule that incorporates into or binds the SEVI-fibrils or an anionic polypeptide supramolecular assembly. The methods can further comprise administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
  • Also provided are pharmaceutical compositions comprising a first agent, which is a semen-derived enhancer of viral infection (SEVI)-binding agent or small molecule (e.g., a hydrophobic molecule that incorporates into ro binds the SEVI-fibrils or an anionic polypeptide supramolecular assembly) as described herein, and a second selected from the group consisting of an anti-viral, an anti-bacterial, and an anti-fungal agent.
  • The details of one or more implementations are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A and 1B are graphs showing semen-derived amyloid fibrils referred to as SEVI (semen-derived enhancer of virus infection) stimulates inflammatory cytokine production by primary human macrophages. FIG. 1A is a graph showing IL-1θ, levels and FIG. 1B is a graph showing TNFI levels in primary human macrophages treated with SEVI or mock treated.
  • FIGS. 2A and 2B show a schematic of a potential mechanism of a microbicide against SEVI (FIG. 2A) and SEVI-binding molecules (2B).
  • FIGS. 3A and 3B show that prostatic acid phosphatase (PAP) 248-286 forms fibrils. PAP248-286 (10 mg/ml in PBS) was agitated at 37° C. and 14,000 RPM. FIG. 3A is a graph showing the results of samples collected at 0 hours, 24 hours, 48 hours, 72 hours, and 96 hours that were subjected to Thioflavin T analysis. FIG. 3B shows images of SEVI-fibrils visualized by electron microscopy. Samples were collected at 72 hours.
  • FIGS. 4A and 4B show that SEVI fibrils enhance HIV-1 infection in vitro. 5×104 CEM 5.25 cells were exposed to infectious HIV-1 (pNL43; 1.2 ng of virus, as determined by p24 ELISA assay) for 2 hours in the presence (10 or 25 μg/mL) or absence of SEVI. FIG. 4A is a graph showing luciferase activity measured in cell lysates at 48 hours post infection. FIG. 4B shows fluorescence microscopy images of GFP at 48 hours.
  • FIGS. 5A and 5B show that the Thioflavin-T analogs BTA-EG4 and BTA-EG6 inhibit SEVI mediated enhancement of HIV infection. CEM 5.25 cells were exposed to infectious HIV-1 (IIIB) for 2 hours in the absence or presence (25 μg/mL) of SEVI fibrils; BTA-EG4 (FIG. 5A) and EG6 (FIG. 5B) was added at concentrations of 5.5, 11 and 16.5 μg/mL. Luciferase activity was measured in cell lysates 72 hrs post-infection. * indicates p value<0.05.
  • FIGS. 6A and 6B show BTA-EG4 and BTA-EG6 inhibit semen mediated enhancement of HIV Infection. HIV-1 IIIB virions were preincubated with 50% semen, with or without increasing concentrations of BTA-EG4 (FIG. 6A) and BTA-EG6 (FIG. 6B). After 10 minutes these stocks were diluted 15 fold into CEM 5.25 cells. Cells were washed after 1 hour and luciferase expression was measured at 48 hours to quantify the extent of infection. * indicates p value<0.05.
  • FIGS. 7A and 7B show BTA-EG4 and BTA-EG6 decrease SEVI enhanced binding of HIV to target cells. HIV-1 (IIIB) virions were pretreated with 10 ug/mL SEVI and added to Jurkat cells with or without increasing concentrations of BTA-EG4 (FIG. 7A) or BTA-EG6 (FIG. 7B). After 90 minutes, cells were washed to remove any unbound virus and bound virions were detected using a p24 ELISA.
  • FIG. 8 shows fluorescence polarization analysis of heparin binding to SEVI fibrils. SEVI was diluted to concentrations ranging from 5 to 100 μg/ml, in the presence of 16 μg/ml of FITC-heparin. Samples were incubated 1 hour at RT, and read at excitation λ=480, and emission λ=535. The graphs show the competitive displacement of bound FITC-heparin from SEVI fibrils. SEVI (100 μg/ml) and FITC-heparin (16 μg/ml) were combined as demonstrated in the left graph. In the right graph, unlabeled heparin was then added, in serial 10-fold dilutions from 3000 to 3 μg/ml.
  • FIGS. 9A and 9B show that fluorescence polarization detects binding of BTA-EG4 (FIG. 9A) and BTA-EG6 (FIG. 9B) to SEVI fibrils. 100 ug/mL of SEVI was mixed with 16 ug/mL FITC-heparin in varying concentrations of BTA-EG4 or BTA-EG6. Samples were incubated 1 hour at room temperature and polarized fluorescence intensities were measured.
  • FIGS. 10A and 10B show BTA-EG4 and EG6 are not toxic to cervical epithelial Cells. The cervical epithelial cell lines A2En (endocervical) (FIG. 10A), 3EC1 and SiHa (FIG. 10B) were treated for 12 hours with BTA-EG4 and BTA-EG6 at concentrations up to 10 times greater than the inhibitory concentration. At 12 hours, viability was measured with Alamar Blue.
  • FIGS. 11A and 11B show BTA-EG4 and BTA-EG6 do not induce cytokine production in cervical epithelial cells. A2En, 3EC1 and SiHa Cells were treated with BTA-EG4 or BTA-EG6 at varying concentrations for 6 hours. At 6 hours, supernatants were collected and cytokine production (IL-1β (FIG. 11A); Mip3α (FIG. 11B); and TNF-α) was determined by ELISA. Representative results from Siha cells are shown.
  • FIG. 12 shows that the thioflavin-T analog BTA-EG6 binds SEVI fibrils. In FIG. 12A shows the chemical structure of ThT and BTA-EG6. FIG. 12B shows that BTA-EG6 binds SEVI fibrils as measured by fluorescence polarization. 100 μg/ml SEVI was mixed with 16 μg/ml FITC-heparin in varying concentrations of BTA-EG6 ranging from 0 to 200 μg/ml. Samples were incubated 1 h at room temperature, and polarized fluorescence intensities were measured. Decreased millipolarization units (mP) indicate a displacement of FITC-heparin from SEVI fibrils due to BTA binding. FIG. 12C shows binding of BTA-EG6 to SEVI fibrils as determined by a centrifugation assay. Briefly, various concentrations of BTA-EG6 in PBS were incubated overnight at room temperature in the presence or absence of SEVI fibrils. After equilibration, each solution was centrifuged, and the supernatants were separated from the pelleted fibrils. The fluorescence of BTA-EG6 was determined from the resuspended pellets in PBS solution. Error bars represent ±S.D. of duplicate measurements. The Kd was determined by fitting the data to a one-site specific binding algorithm: Y=Bmax×X/(Kd+X), where X is the concentration of BTA-EG6, Y is the specific binding fluorescence intensity, and Bmax corresponds to the apparent maximal observable fluorescence upon binding of BTA-EG6 to SEVI fibrils. RFI, relative fluorescence intensity. FIG. 12D shows that BTA-EG6 does not affect the stability of SEVI fibrils. Preformed SEVI fibrils were incubated with increasing concentrations of BTA-EG6 for 3 h. Fibril stability was measured by ThT fluorescence. FIG. 12E shows that BTA-EG6 binding to SEVI inhibits the interaction of SEVI fibrils with the cell surface. Jurkat T cells were incubated with SEVI-biotin for 1 h in the presence or absence of 5.5 μg/ml (low) or 27 μg/ml (high) BTA-EG6. Surface-bound fibrils were detected with SA-FITC and measured by flow cytometry. Results are summarized in Table 1 and are representative of three experiments that were performed with similar results.
  • FIG. 13 shows that BTA-EG6 inhibits SEVI-mediated enhancement of HIV-1 infection. In FIG. 13A, HIV-1 IIIB virions were preincubated with increasing concentrations of BTA-EG6 (0, 5.5, 11, and 22.5 μg/ml) and with or without SEVI (15 μg/ml) as indicated. The samples were then added to CEM-M7 cells. Cells were washed at 2 h, and infection was assayed at 48 h by measuring Tat-driven luciferase expression. Results shown are average values±S.D. of triplicate measurements from one of four independent experiments that yielded equivalent results. * indicates p<0.05 when compared with control cells exposed to HIV-1IIIB+SEVI alone by ANOVA with Tukey's post test. RLU, relative luciferase units; Uninf, uninfected. FIG. 13B is a zoom in of panel A to show data for cells treated with HIV-IIIB virions with and without increasing concentrations of BTA-EG6, in the absence of SEVI. BTA-EG6 had no effect on the infectivity of HIV alone; concentrations of BTA-EG6 are noted above for panel A. FIG. 13C shows the results of CEM-M7 cells infected with HIV-1ADA, as in panel A. FIG. 13D shows that CEM-M7 cells were infected with HIV-1ADA+SEVI with concentrations of BTA-EG6 ranging from 0.4 to 50 μg/ml. An exponential decay curve was then fit to the data and used to calculate the IC50 of the inhibitory effect of BTA-EG6 on SEVI-mediated enhancement of HIV-1 infection. In FIG. 13E, human PBMCs were stimulated with IL-2/PHA and infected with HIV-1BAL and increasing concentrations of BTA-EG6 (0, 5.5, 11, and 22.5_g/ml) with and without SEVI (15 μg/ml). Cells were washed at 3 h, and infection was assayed at 4 days by measuring p24. Results shown are average values±S.D. of triplicate measurements. * indicates p0.01 when compared with control PBMCs exposed to HIV-1ADA_SEVI alone (ANOVA with Tukey's post test).
  • FIG. 14 shows that BTA-EG6 inhibits semen-mediated enhancement of HIV-1 infectivity. FIG. 14A shows HIV-1IIIB virions were preincubated with 50% pooled human semen, with or without increasing concentrations of BTA-EG6 (5.5, 11, and 22.5 μg/ml). After 10 min, these stocks were diluted 15-fold into CEM-M7 cells. Cells were washed after 1 h, and luciferase expression was measured at 48 h to quantify the extent of infection. Results shown are average values±S.D. of triplicate measurements from one of three independent experiments that yielded equivalent results. * indicates p<0.05 when compared with control cells exposed to HIV-1IIIB+semen alone, by ANOVA with Tukey's post test. RLU, relative luciferase units. In FIG. 14B, cells were treated as above but with HIV-1ADA and a 50% concentration of an individual semen sample. *, p<0.05 when compared with control cells exposed to HIV-1ADA+semen alone, by ANOVA with Tukey's post test. FIGS. 14C and D show that BTA-EG6 does not inhibit semen-mediated cytokine release. SiHa cells were treated with pooled human semen for 6 h, with and without 27 μg/ml BTAEG6. At 6 h, IL-8 (C) and MIP-3α (D) production in the supernatants was measured by ELISA. Results shown are average values±S.D. of triplicate measurements from one of three independent experiments that yielded equivalent results. N.S=not significant when compared with cells treated with semen alone (as determined by ANOVA with Tukey's post test).
  • FIG. 15 shows that BTA-EG6 inhibits SEVI-mediated attachment of HIV-1 to the cell surface. In FIG. 15A, HIV-1IIIB virions were pretreated with or without 10 μg/ml SEVI and added to Jurkat cells with or without increasing concentrations of BTA-EG6 (5.5, 11, and 22.5 μg/ml). After 90 min, cells were washed to remove any unbound virus, and bound virions were detected using a p24 ELISA. The data show that BTA-EG6 efficiently inhibited SEVI-mediated enhancement of HIV-1IIIB attachment to Jurkat cells (* indicates p<0.01 for cells treated with SEVI plus 5.5, 11, or 22.5 μg/ml BTA-EG6 versus cells treated with SEVI alone; ANOVA with Tukey's post test). BTA-EG6 had no effect on the binding of HIV-1 virions alone to cells. Uninf, uninfected. In FIG. 15B, Jurkat cells were treated as above using HIV-1ADA (* indicates p<0.01 for cells treated with SEVI plus 11 or 22.5 μg/ml BTA-EG6 versus cells treated with SEVI alone; ANOVA with Tukey's post test). In FIG. 15C, A2En cells were incubated with HIV-1ADA in the presence or absence of 22.5_g/ml BTA-EG6 (* indicates p<0.01 for cells treated with SEVI plus 22.5 μg/ml BTA-EG6 versus cells treated with SEVI alone; ANOVA with Tukey's post test). In FIGS. 15A-C, all results shown are average values±S.D. of triplicate measurements from one of three independent experiments that yielded equivalent results. In FIG. 15D, A2En cells were treated with HIV-1BaL and 15 μg/ml SEVI with or without increasing concentrations of BTA-EG6 (5.5, 11, and 22.5 μg/ml). At 24 h, supernatants were collected and analyzed by ELISA for the presence of IL-8. (* indicates p<0.01 for cells treated with SEVI plus 11 or 22.5 μg/ml BTA-EG6 versus cells treated with SEVI alone; ANOVA with Tukey's post test).
  • FIG. 16 shows that BTA-EG6 is not toxic to cervical cells. In FIG. 16A, the cervical endothelial cell lines A2En (endocervical), 3EC1 (ectocervical), and SiHa were treated for 12 h with BTA-EG6 at concentrations up to 10 times greater than the IC50. Control cultures were treated with nonoxynol-9 (non-9) at 0.1% final concentration as a positive control for induction of cell death. At 12 h, viability was measured by resazurin cytotoxicity assay (AlamarBlue assay). Representative results from A2En cells are shown; results from 3EC1 and SiHa cells were very similar. In FIGS. 16B and C, BTA-EG6 does not induce inflammatory chemokine production in cervical epithelial cells. A2En, 3EC1, and SiHa Cells were treated with BTA-EG6 at varying concentrations for 6 h; control cultures were treated with a well defined TLR2/6 agonist, FSL1 (a synthetic diacylated lipoprotein derived from M. salivarium) at 0.1 μg/ml final concentration as a positive control for chemokine induction. At 6 h, supernatants were collected, and production of Mip-3α (B) and IL-8 (C) was determined by ELISA. Representative results from A2En cells are shown; results from 3EC1 and SiHa cells were very similar. In FIGS. 16A-C, all results shown are average values±S.D. of triplicate measurements from one of three independent experiments that yielded equivalent results. No significant difference (p>0.05) was noted between control cultures treated with PBS and those treated with the highest dose (66 μg/ml) of BTA-EG6, as determined by ANOVA with Tukey's post test.
  • FIGS. 17A and B show levels of bound virons using an HIV-1 p24 antigen capture assay with HIV-1 IIIB virions pretreated with 15 μg/ml SEVI and added to 5×104 A2En cells (immortalized primary human endocervical cells) (A) or to Jurkat T cells (a CD4+ human T cell line) (B) in the presence or absence of test compounds (at a final concentration of 25 μM).
  • FIG. 18A shows a schematic of binding of an amyloid-binding ligand, like benzothiazole aniline (BTA), in monomeric (left panel) or oligomeric (right panel) form. FIG. 18B shows the structure of a benzothiazole aniline (BTA)-based monomer (1), dimer (2), trimer (3), tetramer (4), and pentamer (5). The structure of BTA moiety is given and is represented as simple red ovals in molecules 1-5 for clarity.
  • DETAILED DESCRIPTION
  • The majority of sexually transmitted infections are acquired through unprotected sexual relations, that is, sexual intercourse in the absence of a barrier such as a condom. For example, sexual transmission of HIV can occur when HIV-containing secretions, e.g., seminal or vaginal fluid, of one partner come into contact with the genital, oral, or rectal mucous membranes of another. The epithelial cells of the mucous membranes act, at least in part, as a barrier to viral penetration. HIV can cross the epithelial barrier either by capture by intra-epithelial dendritic cells that convey the virus to target cells deeper in the mucosa or through regions of damaged epithelium resulting from traumatic injury or lesions caused by sexually transmitted diseases. Once the virus has breached the epithelial membrane, the infection spreads among cells of the immune system, including, for example, CD4+ T cells, macrophages and dendritic cells. Ultimately, the virus disseminates via the lymphatic system and the blood to spleen, brain, liver, and lungs. The efficiency of sexual transmission of HIV depends on many factors, including, for example, host factors in both the transmitting partner and the recipient. Seminal fluid contains a number of factors, for example, semen fibrils, amines such as spermine, spermidine, putrescine and cadavarine, as well as nutrients and enzymes that protect the virus from the acidic environment of the vaginal tract and that enhance sexual transmission of HIV.
  • Cationic polymers enhance retrovirus transduction by neutralizing the electrostatic repulsion between the virus and cell surface and allowing many virus particles to aggregate onto a single surface enhancing the effective multiplicity of infection. As described herein, semen fibrils (e.g., prostatic acid phosphatase (PAP) fibrils) work in a similar manner since semen fibrils are highly cationic. As described herein, and without meaning to be limited by theory, interfering with the binding of infectious agents such as viruses to semen fibrils reduces the risk of sexually transmitted infections. Immunization against semen-derived amyloid fibrils or precursor forms of such fibrils (e.g., peptide oligomers) will not result in autoimmune reactions against wild-type PAP since the PAP-derived amyloid fibrils and their precursor molecules possess unique conformational attributes that distinguish them from the native PAP protein. Further, PAP has been shown to be a safe vaccine antigen in the context of immunization for prostate cancer. Thus, immunization with short linear peptides derived from PAP is safe.
  • An amyloid-binding small molecule is an efficient inhibitor of SEVI- and semen-mediated enhancement of HIV infectivity. BTA-EG6 binds to the SEVI fibrils and interferes with their ability to enhance HIV infectivity. Importantly, BTA-EG6 did not have any direct inhibitory effects on the infectivity of HIV-1 alone.
  • Provided herein are methods of treating or preventing a sexually transmitted infection in a subject. The methods, optionally, comprise identifying a subject with or at risk of developing a sexually transmitted infection and administering to the subject a semen-derived enhancer of viral infection (SEVI)-binding agent, wherein the agent comprises a compound represented by Formula I:
  • Figure US20130157924A1-20130620-C00001
  • and pharmaceutically acceptable salts and prodrugs thereof. The agent can, for example, bind and prevent the ability of SEVI-fibrils or prefibrillar forms of SEVI from enhancing a sexually transmitted infection in the subject. Optionally, the methods further comprise administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
  • In Formula I, R1, R2, R3, R4, R5, R6, R7, and R8 are each independently selected from hydrogen, halogen, hydroxyl, trifluoromethyl, substituted or unsubstituted thio, substituted or unsubstituted alkoxyl, substituted or unsubstituted aryloxyl, substituted or unsubstituted amino, substituted or unsubstituted C1-12 alkyl, substituted or unsubstituted C2-12 alkenyl, substituted or unsubstituted C2-12 alkynyl, substituted or unsubstituted C1-12 heteroalkyl, substituted or unsubstituted C2-12 heteroalkenyl, substituted or unsubstituted C2-12 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. In some examples, R3 is methyl and R1, R2, R4, R5, R6, R7, and R8 are hydrogen.
  • Also, in Formula I, R9 and R10 are each independently selected from hydrogen, substituted or unsubstituted C1-20 alkyl, substituted or unsubstituted C2-20 alkenyl, substituted or unsubstituted C2-20 alkynyl, substituted or unsubstituted C1-20 heteroalkyl, substituted or unsubstituted C2-20 heteroalkenyl, substituted or unsubstituted C2-20 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. R9 and R10 can combine with the adjacent N to form a heterocycle. In some examples, R10 is hydrogen. In some examples, R9 is the Structure I-A:
  • Figure US20130157924A1-20130620-C00002
  • In Structure I-A, n is an integer from 0 to 20. In some examples of Structure I-A, n is 4 or 6.
  • In Formula I, adjacent R groups on the phenyl ring (i.e., R1, R2, R3, and R4; R5 and R6; and R7 and R8) can be combined to form substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, substituted or unsubstituted cycloalkynyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, or substituted or unsubstituted heterocycloalkynyl groups. For example, R1 can be a formamide group and R2 can be an ethylene group that combine to form a pyridinone group. Other adjacent R groups include the combinations of R2 and R3, R3 and R4, R5 and R6, and R7 and R8.
  • Specific examples of Formula I are as follows:
  • Figure US20130157924A1-20130620-C00003
  • As used herein, the terms alkyl, alkenyl, and alkynyl include straight- and branched-chain monovalent substituents. Examples include methyl, ethyl, isobutyl, 3-butynyl, and the like. Ranges of these groups useful with the compounds and methods described herein include C1-C20 alkyl, C2-C20 alkenyl, and C2-C20 alkynyl. Additional ranges of these groups useful with the compounds and methods described herein include C1-C12 alkyl, C2-C12 alkenyl, C2-C12 alkynyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C4 alkyl, C2-C4 alkenyl, and C2-C4 alkynyl.
  • Heteroalkyl, heteroalkenyl, and heteroalkynyl are defined similarly as alkyl, alkenyl, and alkynyl, but can contain O, S, or N heteroatoms or combinations thereof within the backbone. Ranges of these groups useful with the compounds and methods described herein include C1-C20 heteroalkyl, C2-C20 heteroalkenyl, and C2-C20 heteroalkynyl. Additional ranges of these groups useful with the compounds and methods described herein include C1-C12 heteroalkyl, C2-C12 heteroalkenyl, C2-C12 heteroalkynyl, C1-C6 heteroalkyl, C2-C6 heteroalkenyl, C2-C6 heteroalkynyl, C1-C4 heteroalkyl, C2-C4 heteroalkenyl, and C2-C4 heteroalkynyl.
  • The terms cycloalkyl, cycloalkenyl, and cycloalkynyl include cyclic alkyl groups having a single cyclic ring or multiple condensed rings. Examples include cyclohexyl, cyclopentylethyl, and adamantanyl. Ranges of these groups useful with the compounds and methods described herein include C3-C20 cycloalkyl, C3-C20 cycloalkenyl, and C3-C20 cycloalkynyl. Additional ranges of these groups useful with the compounds and methods described herein include C5-C12 cycloalkyl, C5-C12 cycloalkenyl, C5-C12 cycloalkynyl, C5-C6 cycloalkyl, C5-C6 cycloalkenyl, and C5-C6 cycloalkynyl.
  • The terms heterocycloalkyl, heterocycloalkenyl, and heterocycloalkynyl are defined similarly as cycloalkyl, cycloalkenyl, and cycloalkynyl, but can contain O, S, or N heteroatoms or combinations thereof within the cyclic backbone. Ranges of these groups useful with the compounds and methods described herein include C3-C20 heterocycloalkyl, C3-C20 heterocycloalkenyl, and C3-C20 heterocycloalkynyl. Additional ranges of these groups useful with the compounds and methods described herein include C5-C12 heterocycloalkyl, C5-C12 heterocycloalkenyl, C5-C12 heterocycloalkynyl, C5-C6 heterocycloalkyl, C5-C6 heterocycloalkenyl, and C5-C6 heterocycloalkynyl.
  • Aryl molecules include, for example, cyclic hydrocarbons that incorporate one or more planar sets of, typically, six carbon atoms that are connected by delocalized electrons numbering the same as if they consisted of alternating single and double covalent bonds. An example of an aryl molecule is benzene. Heteroaryl molecules include substitutions along their main cyclic chain of atoms such as O, N, or S. When heteroatoms are introduced, a set of five atoms, e.g., four carbon and a heteroatom, can create an aromatic system. Examples of heteroaryl molecules include furan, pyrrole, thiophene, imadazole, oxazole, pyridine, pyrazole, and pyrazine. Aryl and heteroaryl molecules can also include additional fused rings, for example, benzofuran, indole, benzothiophene, naphthalene, anthracene, and quinoline.
  • The alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl molecules used herein can be substituted or unsubstituted. As used herein, the term substituted includes the addition of an alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl group to a position attached to the main chain of the alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl, e.g., the replacement of a hydrogen by one of these molecules. Examples of substitution groups include, but are not limited to, hydroxyl, halogen (e.g., F, Br, Cl, or I), and carboxyl groups. Conversely, as used herein, the term unsubstituted indicates the alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkynyl, heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl has a full complement of hydrogens, i.e., commensurate with its saturation level, with no substitutions, e.g., linear decane (—(CH2)9—CH3).
  • The compounds described herein can be prepared in a variety of ways known to one skilled in the art of organic synthesis or variations thereon as appreciated by those skilled in the art. The compounds described herein can be prepared from readily available starting materials. Optimum reaction conditions may vary with the particular reactants or solvents used, but such conditions can be determined by one skilled in the art.
  • Variations on the Formula I include the addition, subtraction, or movement of the various constituents as described for each compound. Similarly, when one or more chiral centers are present in a molecule, the chirality of the molecule can be changed. The compounds described herein can be isolated in pure form or as a mixture of isomers. Additionally, compound synthesis can involve the protection and deprotection of various chemical groups. The use of protection and deprotection, and the selection of appropriate protecting groups can be determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4th Ed., Wiley & Sons, 2006, which is incorporated herein by reference in its entirety.
  • Reactions to produce the compounds described herein can be carried out in solvents, which can be selected by one of skill in the art of organic synthesis.
  • Solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products under the conditions at which the reactions are carried out, i.e., temperature and pressure. Reactions can be carried out in one solvent or a mixture of more than one solvent. Product or intermediate formation can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • Provided herein are methods of treating or preventing a sexually transmitted infection in a subject. The methods comprise administering to the subject the compounds described herein, wherein the compounds bind the SEVI-fibrils to inhibit the ability of the SEVI-fibrils to enhance a sexually transmitted infection. Compounds contained within International Publication No. WO 2007/011834 are also contemplated herein for use in methods of treating or preventing a sexually transmitted infection.
  • Also provided are methods of treating or preventing a sexually transmitted infection in a subject. The methods, optionally, comprise identifying a subject with or at risk of developing a sexually transmitted infection and administering to the subject a semen-derived enhancer of viral infection (SEVI)-binding small molecule. The methods can further comprise administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
  • The SEVI-binding small molecule can, for example, comprise a hydrophobic molecule, wherein the hydrophobic molecule incorporates into and binds the SEVI-fibrils. SEVI-fibrils are formed as a result of ahydrophobic interactions between component monomer polypeptides. Without meaning to be limited by theory, it is expected that exogenous hydrophobic molecules, such as hydrophobic polypeptides, can be incorporated into and bind the SEVI-fibrils, thus inhibiting the ability of the SEVI-fibrils to interact with the infectious agent causing the sexually transmitted infection. Examples of such hydrophobic molecules include alkanes, oils, fats, and greasy substances in general.
  • The SEVI-binding small molecule can, for example, comprise an anionic polypeptide supramolecular assembly. Optionally, the anionic supramolecular assembly is water-soluble. Optionally, the anionic supramolecular assembly comprises a soluble hydrogel and other supramolecular assemblies derived from an Ac-(XEXE)n-NH2 (SEQ ID NO:14) polypeptide and related polypeptides. Water-soluble supramolecular assemblies derived from self-assembling anionic polypeptides can, for example, bind to the cationic SEVI fibrils and inhibit interactions between the SEVI-fibrils and the infectious agents. An example of a soluble hydrogel is the PuraMatrix hydrogel. The PuraMatrix hydrogel comprises a (VKVK)n polypeptide fibrillar hydrogel that is not toxic.
  • The SEVI-binding small molecules can further comprise a bulky side chain, a negatively charged side chain, a coupled moiety, and an anti-viral molecule. A bulky side chain can, for example, comprise a poly-ethylene glycol (PEG) molecule. An anti-viral molecule can, for example, comprise a pradimicin A or AZT molecule.
  • Also provided are methods of screening for an agent that is capable of binding SEVI-fibrils. Methods of screening for agents that are capable of binding SEVI-fibrils include the steps of providing the agent to be screened, contacting the agent with the SEVI-fibrils, and determining whether the agent to be screened binds the SEVI-fibrils. Binding can be determined, for example, by selecting an assay from the group consisting of a coimmunoprecipitation assay, a colocalization assay, or a fluorescence polarizing assay, as described below. The assays are known in the art, e.g., see Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd Ed., Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (2001); Dickson, Methods Mol. Biol. 461:735-44 (2008); Nickels, Methods 47(1):53-62 (2009); and Zinchuk et al., Acta Histochem. Cytochem. 40(4):101-11 (2007).
  • The SEVI-binding agents, SEVI-binding small molecules, anti-viral agents, anti-bacterial agents, anti-fungal agents described herein or derivatives thereof can be provided in a pharmaceutical composition. Depending on the intended mode of administration, the pharmaceutical composition can be in the form of solid, semi-solid or liquid dosage forms, such as, for example, tablets, suppositories, pills, capsules, powders, liquids, or suspensions, preferably in unit dosage form suitable for single administration of a precise dosage. The compositions will include a therapeutically effective amount of the compound described herein or derivatives thereof in combination with a pharmaceutically acceptable carrier and, in addition, may include other medicinal agents, pharmaceutical agents, carriers, or diluents. By pharmaceutically acceptable is meant a material that is not biologically or otherwise undesirable, which can be administered to an individual along with the selected compound without causing unacceptable biological effects or interacting in a deleterious manner with the other components of the pharmaceutical composition in which it is contained.
  • Such pharmaceutical compositions are optionally, provided in the form of contraceptives or contraceptive agents, such as condoms or spermicides, or lubricants.
  • As used herein, the term carrier encompasses any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, lipid, stabilizer, or other material well known in the art for use in pharmaceutical formulations. The choice of a carrier for use in a composition will depend upon the intended route of administration for the composition. The preparation of pharmaceutically acceptable carriers and formulations containing these materials is described in, e.g., Remington's Pharmaceutical Sciences, 21st Edition, ed. University of the Sciences in Philadelphia, Lippincott, Williams & Wilkins, Philadelphia Pa., 2005. Examples of physiologically acceptable carriers include buffers such as phosphate buffers, citrate buffer, and buffers with other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN® (ICI, Inc.; Bridgewater, N.J.), polyethylene glycol (PEG), and PLURONICS™ (BASF; Florham Park, N.J.).
  • Compositions containing the compound described herein or derivatives thereof suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be promoted by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. Isotonic agents, for example, sugars, sodium chloride, and the like may also be included. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Solid dosage forms for oral administration of the compounds described herein or derivatives thereof include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the compounds described herein or derivatives thereof is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders, as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate, (e) solution retarders, as for example, paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol, and glycerol monostearate, (h) adsorbents, as for example, kaolin and bentonite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethyleneglycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others known in the art. They may contain opacifying agents and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes. The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration of the compounds described herein or derivatives thereof include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents, and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols, and fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • Besides such inert diluents, the composition can also include additional agents, such as wetting, emulsifying, suspending, sweetening, flavoring, or perfuming agents.
  • Suspensions, in addition to the active compounds, may contain additional agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • Compositions of the compounds described herein or derivatives thereof for rectal administrations are preferably suppositories, which can be prepared by mixing the compounds with suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
  • Dosage forms for topical administration of the compounds described herein or derivatives thereof include ointments, powders, sprays, gels and the like. The compounds described herein or derivatives thereof are admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as may be required.
  • The term pharmaceutically acceptable salt as used herein refers to those salts of the compound described herein or derivatives thereof that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds described herein. The term salts refers to the relatively non-toxic, inorganic and organic acid addition salts of the compounds described herein. These salts can be prepared in situ during the isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate, methane sulphonate, and laurylsulphonate salts, and the like. These may include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. (See S. M. Barge et al., J. Pharm. Sci. (1977) 66, 1, which is incorporated herein by reference in its entirety, at least, for compositions taught herein.)
  • The active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. Those of skill in the art will understand that the specific dose level and frequency of dosage for any particular subject may be varied, and it will be understood that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and response of the individual subject, the severity of the subject's symptoms, and the like.
  • Methods of Treatment or Prevention
  • The compositions described herein are useful for preventing or reducing the transmission of sexually transmitted infections (STIs). For example, administering a SEVI-binding agent or SEVI-binding small molecule to a subject interferes with the binding of infectious agents to the semen fibrils. Such binding interferes with the infection-enhancing activity of the semen fibrils and prevents or reduces the risk of STIs. The compositions are useful for treatment prior to, during, or after infection. Treatment can completely or partially abolish some or all of the signs and symptoms of transmission of the infection and reduce the likelihood that the treated subject will subsequently develop symptoms of an STI or will delay the onset of symptoms. Thus, for example, treatment can prevent, reduce or delay viral transmission, e.g., HIV transmission.
  • STIs are infections that can be transferred from one subject to another through sexual contact. In general, STIs are caused by microorganisms that are transmitted via semen, vaginal secretions or blood during sexual contact or by microorganisms that survive on the skin and mucous membranes of the genital area. Sexual contact can include sexual intercourse (vaginal and anal), oral-genital contact, and the use of sexual toys, such as vibrators. Microorganisms transmitted via sexual contact can include, for example, viruses, e.g., HIV, human papilloma virus (HPV), herpesviruses, hepatitis B, and C and cytomegalovirus (CMV); bacteria, e.g., infectious agents responsible for gonorrhea (Neisseria gonorrhoeae); syphilis (Treponema pallidum); chancroid (Haemophilus ducreyi); donovanosis (Granuloma inguinale or Calymmatobacterium granulomatis); lymphogranuloma venereum (LGV) (Chlamydia trachomatis); non-gonococcal urethritis (NGU) (Ureaplasma urealyticum or Mycoplasma hominis); bacterial vaginosis and Staphylococcus aureus; protozoa, e.g., infectious agents responsible for trichommoniasis (Trichomonas vaginalis).
  • Symptoms of STIs can vary and often the infected subject has no symptoms. However, an asymptomatic subject may be able to pass the disease to a sexual partner. Common symptoms of STI's include, but are not limited to, urethral discharge, genital ulcers, inguinal swellings, scrotal swelling, vaginal discharge, lower abdominal pain, fever, lymphadenopathy (swollen lymph nodes), pharyngitis (sore throat), rash, myalgia (muscle pain), malaise, and mouth and esophageal sores. Both symptomatic and asymptomatic infections can lead to the development of more serious conditions, including AIDS, pelvic inflammatory disease, infertility and tubal (ectopic) pregnancy, genital warts, cervical and other genital cancers.
  • The compositions and methods are applicable to the transmission of infections by any type of HIV, e.g., HIV-1 and HIV-2. The compositions can be administered to both men and women. The compositions are suitable for a subject who is not infected with HIV, but is at risk for sexually transmitted infection. Subjects who may be at increased risk of becoming infected through sexual contact include those who have unprotected sex, i.e., do not use condoms during sexual intercourse; have multiple sex partners; males who have sexual intercourse with other men; those who have high-risk partner(s), i.e., the sexual partner has multiple sex partners, is a man who has sex with other men, or is an intravenous drug user; or those who have or have recently had a sexually transmitted disease, e.g., syphilis, gonorrhea of chlamydia.
  • The compositions are also useful in an infected subject, e.g., a subject who has an HIV infection, to reduce the transmission to an uninfected partner. The compositions can be administered to a subject at any stage in the course of HIV infection.
  • The efficacy of the compositions can be monitored according to standard methods in the art for assessing HIV status, including measuring the level of HIV, using for example a PCR assay, in a clinical sample, e.g., a blood sample, measuring the level of anti-HIV antibodies, using for example, an ELISA or immunoblotting assay, in a clinical sample, e.g., a blood sample, and by monitoring the levels of CD4+ T cells in a clinical sample.
  • The compositions can be administered in conjunction with other therapeutic or prophylactic modalities to an individual in need of treatment. For example, the compositions may be administered to a subject who practices “safe sex”, i.e., a subject who wears a condom during sexual intercourse or has sexual intercourse with a partner who wears a condom. The condom can be disguised to contain or be coated with the therapeutic agent. The compositions can also be administered in conjunction with other therapies for treating HIV infection, such as standard small molecule pharmaceutical agents, topical microbicides, biopharmaceuticals (e.g., antibodies or antibody-related immunotherapies, siRNAs, shRNAs, antisense oligonucleotides and other RNA inhibitory molecules, microRNAs, and peptide therapeutics), surgery, or in conjunction with any medical devices that may be used to assist the subject. Standard therapy for HIV infection includes highly active antiretroviral therapy, or HAART. Typically, HAART includes a combination (or “cocktail”) of drugs belonging to at least two classes of antiretroviral agents, e.g., a nucleoside analogue reverse transcriptase inhibitors (NARTIs or NRTIs), a non-nucleoside reverse transcriptase inhibitor and a protease inhibitor. Nucleoside reverse transcriptase inhibitors include, for example: AZT (ZDV, zidovudine, Retrovir), ddI (didanosine, Videx), d4T (stavudine, Zerit), 3TC (lamivudine, Epivir), Abacavir (Ziagen), Tenofovir (Viread), Combivir (AZT/3TC combination), Trizivir (AZT/3TC/Abacavir combination), Emtricitabine (FTC, Emtriva), Epzicom (3TC/abacavir combination) and Truvada (tenofovir/emtricitabine combination). Non-nucleoside reverse transcriptase inhibitors (NNRTIs) include, for example: Nevirapine (NVP, Viramune), Delavirdine (DLV, Rescriptor), Efavirenz (EFV, Sustiva, Stocrin) and Etravirine (ETV, Intelence). Protease inhibitors include, for example: Saquinavir (SQV, Invirase), Indinavir (IDV, Crixivan), Ritonavir (RTV, Norvir), Nelfinavir (NFV, Viracept), Amprenavir (APV, Agenerase), Lopinavir (LPV, Kaletra, Aluvia), Atazanavir (ATV, Reyataz), Fosamprenavir (FPV, Lexiva), Tipranavir (TPV, Aptivus) and Darunavir (DRV, Prezista). Other anti-HIV drugs include fusion and attachment inhibitors, including, for example, Enfuvirtide (Fuzeon or T-20) and Maraviroc (MVC, Selzentry, Celsentri); and integrase inhibitor, including for example, Raltegravir (RGV, Isentress). Optionally, the compositions can be incorporated into standard barrier prophylatics, for example male and female condoms.
  • The duration of treatment with any composition provided herein can be any length of time from as short as one day to as long as the life span of the host (e.g., many years). For example, the composition can be administered once a week (for, for example, 4 weeks to many months or years); once a month (for, for example, three to twelve months or for many years); or once a year for a period of 5 years, ten years, or longer. It is also noted that the frequency of treatment can be variable. For example, the compositions can be administered once (or twice, three times, etc.) daily, weekly, monthly, or yearly.
  • An effective amount of any composition provided herein can be administered to an individual in need of treatment. The term “effective” as used herein refers to any amount that induces a desired response while not inducing significant toxicity in the patient. Such an amount can be determined by assessing a patient's response after administration of a known amount of a particular composition. In addition, the level of toxicity, if any, can be determined by assessing a patient's clinical symptoms before and after administering a known amount of a particular composition. It is noted that the effective amount of a particular composition administered to a patient can be adjusted according to a desired outcome as well as the patient's response and level of toxicity. Significant toxicity can vary for each particular patient and depends on multiple factors including, without limitation, the patient's disease state, age, and tolerance to side effects.
  • In addition, clinical methods that can assess the degree of a particular disease state can be used to determine if a response is induced. For example blood or laboratory tests may be administered to determine HIV titers before, during and after a course of treatment. The particular methods used to evaluate a response will depend upon the nature of the patient's disorder, the patient's age, and sex, other drugs being administered, and the judgment of the attending clinician.
  • Kits
  • The compositions described herein can also be assembled in kits, together with instructions for use and/or containers, means for administration of the composition, and the like. For example, the kits can include measured amounts of a pharmaceutically acceptable composition including the compounds described herein, and the anti-viral, anti-bacterial, or anti-fungal agents described herein. The instructions for use can be conveyed by any suitable media. For example, they can be printed on a paper insert in one or more languages or supplied audibly or visually (e.g., on a compact disc). The packaging materials can include vials, packets, or intravenous bags, and the kit can also include instruments useful in administration, such as needles, syringes, tubing, condoms, catheters, bandages, and tape. Preferably, the components of the kit are sterile and suitable for immediate use. The invention encompasses kits, however, that include concentrated formulations and/or materials that may require sterilization prior to use.
  • Semen Fibrils
  • The semen fibrils comprise fibrillary aggregates derived from polypeptides in seminal fluid. The fibrillary aggregates can be insoluble fibrous protein aggregates that are generally characterized by a cross-beta sheet quaternary structure; i.e., a monomeric unit contributes a beta strand to a beta sheet, which spans across more than one molecule. The fibrils can be identified using a variety of assays, including fluorescent dyes, e.g., thioflavin T binding, Congo red staining, stain polarimetry, circular dichroism, FTIR or X-ray diffraction analysis. X-ray diffraction analysis reveals characteristic scattering diffraction signals produced at 4.7 and 10.6 Ångstroms (0.47 nm and 1.06 nm), corresponding to the interstrand and stacking distances in beta sheets. The stacks of beta sheet are short and traverse the breadth of the amyloid fibril; the length of the fibril is built by aligned strands.
  • Semen fibrils can form from semen fibrillary polypeptides or oligomers thereof. A semen fibrillary polypeptide can be a fibril forming fragment of prostatic acid phosphatase (PAP), a protein produced by the prostate and secreted into semen. PAP (also known as ACP-3 or prostatic acid phosphatase precursor 3, ACP3, ACPP or EC 3.1.3.2) is the prostate-specific form of one of five ubiquitous acid phosphatase isozymes that catalyze the conversion of orthophosphoric monoester to alcohol and orthophosphate. PAP is over 100 times more abundant in the prostate that in other tissue types. The cDNA and amino acid sequences encoding a representative human PAP polypeptide (Genbank number NM001099 [gi:161377405] and NP001090 [gi:6382064]) are shown as SEQ ID NOs: 1 and 2, respectively. Other amino acid sequences that have been identified for PAP include, without limitation, BAD89417.1, [gi:58737017]; AAB60640, [gi:515997]; AAA60021, [gi:189613]; and NP 064457, [gi:9910502]. Additional amino acid modifications may include PAP-derived sequence derivatives with extensive stretches of hydrophobicity and an associated predilection for fibril formation. The amino acid sequence of human PAP is 386 residues in length; the active form of the enzyme is a homodimer. A peptide corresponding to the amino acid sequence from about residue 248 to about residue 286 in human PAP, i.e.,
  • (SEQ ID NO: 3)
    YGIHKQKEKSRLQGGVLVNEILNHMKRATQIPSYKKLIMY

    forms fibrils that enhance the transmission of HIV.
  • DEFINITIONS
  • As used throughout, subject can be a vertebrate, more specifically a mammal (e.g. a human, horse, cat, dog, cow, pig, sheep, goat, mouse, rabbit, rat, and guinea pig), birds, reptiles, amphibians, fish, and any other animal. The term does not denote a particular age or sex. Thus, adult and newborn subjects, whether male or female, are intended to be covered. As used herein, patient or subject may be used interchangeably and can refer to a subject with a sexually transmitted infection. The term patient or subject includes human and veterinary subjects.
  • As used herein the terms treatment, treat, or treating refers to a method of reducing the effects of a sexually transmitted infection or a symptom of the sexually transmitted infection as described above. Thus in the disclosed method, treatment can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% reduction in the severity of a sexually transmitted infection or a symptom of the sexually transmitted infection. For example, a method for treating a sexually transmitted infection is considered to be a treatment if there is a 10% reduction in one or more symptoms of the infection in a subject as compared to a control. Thus, the reduction can be a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or any percent reduction in between 10% and 100% as compared to native or control levels. It is understood that treatment does not necessarily refer to a cure or complete ablation of the infection or symptoms of the infection.
  • As used herein, the terms prevent, preventing, and prevention of a sexually transmitted infection as described above refers to an action, for example, administration of a therapeutic agent, that occurs before or at about the same time a subject begins to show one or more symptoms of the disease or disorder, which inhibits or delays onset or exacerbation of one or more symptoms of the infection. As used herein, references to decreasing, reducing, or inhibiting include a change of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater as compared to a control level. Such terms can include but do not necessarily include complete elimination.
  • Optional or optionally means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of skill in the art to which the disclosed method and compositions belong. Publications cited herein and the material for which they are cited are hereby specifically incorporated by reference in their entireties.
  • It is understood that the disclosed method and compositions are not limited to the particular methodology, protocols, and reagents described as these may vary.
  • Disclosed are materials, compositions, and components that can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed methods and compositions. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutations of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a method is disclosed and discussed and a number of modifications that can be made to a number of molecules including the method are discussed, each and every combination and permutation of the method, and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. Likewise, any subset or combination of these is also specifically contemplated and disclosed. This concept applies to all aspects of this disclosure including, but not limited to, steps in methods using the disclosed compositions. Thus, if there are a variety of additional steps that can be performed, it is understood that each of these additional steps can be performed with any specific method steps or combination of method steps of the disclosed methods, and that each such combination or subset of combinations is specifically contemplated and should be considered disclosed.
  • Publications cited herein and the material for which they are cited are hereby specifically incorporated by reference in their entireties.
  • EXAMPLES Example 1 Administration of Semen Enhancer of Viral Infection (SEVI) to Primary Human Macrophages Stimulates Inflammatory Cytokine Production
  • Primary human macrophages were prepared from whole blood by Lymphoprep (Accurate Chemical & Scientific; Westbury, N.Y.) density-gradient centrifugation followed by positive selection with CD 14+ microbeads (Miltenyi Biotec; Bergisch Gladbach, Germany). The cells were plated in 48-well plates at a concentration of 5×105 cells/mL and differentiated using RPMI-1640 supplemented with 20% fetal bovine serum (FBS) and 5 ng/mL granulocyte macrophage-colony stimulating factor (GM-CSF). After 4 days, the cells were maintained with RPMI-1640 with 20% FBS.
  • After 7 days, the primary human macrophages were stimulated with either LPS (100 ng/mL), SEVI (10 mM), or both. Cell culture supernatants were collected at 0, 4, and 24-hour timepoints and measurements of TNFα and IL-1β were determined by ELISA. Briefly, 96-well plates were coated with 100 μL/well of capture antibody in coating buffer (eBioscience, Inc.; San Diego, Calif.) and incubated overnight at 4° C. The wells were washed with phosphate buffered saline (PBS) with 0.05% Tween-20 and blocked for 1 hour with 300 μL/well assay diluent (eBioscience, Inc.). 100 μL of the samples (cell culture supernatants) or standards (eBioscience, Inc.) were incubated for 2 hours at room temperature. After washing the wells, 100 μL/well of biotin-conjugated anti-human IL-1β or biotin-conjugated anti-human TNFα detection antibody (eBioscience, Inc.) was added for 1 hour, followed by 100 μL of Streptavidin-HRP (eBioscience, Inc.) for 30 minutes. The wells were developed with TMB (eBioscience, Inc.) and the reaction was stopped with 2N H2SO4. Optical density was read at 450 nm with a SpectraCount plate reader (Packard Instrument Company; Meriden, Conn.), and the cytokine levels were then calculated by extrapolation to a standard curve generated using known amounts of recombinant IL-1β and TNFα.
  • The results are shown in FIG. 1. Addition of SEVI to primary human macrophages as compared to a control elicits an increase in inflammatory cytokine production as evidenced by the increase in IL-1β and TNFα. The results are presented as mean plus or minus the standard error of the mean (SEM) for three independent cell replicates (obtained from a single unit of human blood).
  • Example 2 Identification of Compounds that Bind SEVI and Inhibit SEVI's Effects on HIV Infection Synthesis of BTA-EG6
  • BTA-EG6 was synthesized as described previously (Inbar, P., Li, C. Q., Takayama, S. A., Bautista, M. R., and Yang, J. (2006) Chembiochem 7, 1563-1566).
  • Cell Culture
  • CEM-M7 (a gift from N. Landau, New York University, New York, N.Y.) and Jurkat cells were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum, penicillin (50 units/ml), and streptomycin (50 μg/ml). SiHa cells were cultured in DMEM medium supplemented with 10% fetal bovine serum, penicillin (50 units/ml), and streptomycin (50 μg/ml). A2En cells (a gift from S. Greene, Louisiana State University Health Sciences Center, New Orleans, La.), and 3EC1 cells (a gift from R. Pyles, University of Texas Medical Branch, Galveston, Tex.) were cultured in keratinocyte serum-free medium (Invitrogen, Carlsbad, Calif.) supplemented with bovine pituitary extract (50 mg/liter), recombinant epidermal growth factor (5 μg/liter), CaCl2 (44.1 mg/liter), and Primocin (0.1 mg/ml). PBMCs were isolated from whole blood by Lymphoprep density gradient centrifugation. PBMCs were stimulated for 48 h in RPMI 1640 medium supplemented with 5% human IL-2 (ZeptoMetrix, Buffalo, N.Y.), 5 μg/ml PHA (Sigma, St. Louis, Mo.), 20% fetal bovine serum, penicillin (50 units/ml), and streptomycin (50 μg/ml).
  • SEVI and Semen
  • PAP248-286 and biotinylated PAP248-286, in which a biotin was added to the amino terminus of the peptide, was synthesized and dissolved in PBS at a concentration of 10 mg/ml. Fibrils were formed by agitation in an Eppendorf Thermomixer at 1400 rpm (Eppendorf, Hauppauge, N.Y.) and 37° C. for 72 h. Semen samples were obtained from the Strong Fertility Center (Rochester, N.Y.) and Fairfax CryoBank (Fairfax, Va.). Samples were pooled, aliquoted, and stored at −80° C.
  • Fluorescence Polarization
  • 100 μg/ml SEVI was mixed with 16 μg/ml FITC-heparin and concentrations of BTA-EG6 ranging from 0 to 200 μg/ml. Samples were incubated 1 h at room temperature and read on a PerkinElmer Life Sciences Envision 2012 multilabel reader (PerkinElmer, Waltham, Mass.) at an excitation λ=480 nm and emission λ=535 nm. The horizontal and vertical polarized fluorescence intensities were recorded, and the calculated polarization was determined in millipolarization units.
  • Determination of the Binding Affinity of BTA-EG6 and SEVI Fibrils
  • Binding of BTA-EG6 to SEVI fibrils was measured according to the centrifugation assay described by Levine (LeVine, H., 3rd (2005) Amyloid 12: 5-14) for BTA-1 to Aβ fibrils. Briefly, 200 μl of various concentrations of BTA-EG6 in PBS were incubated in the presence or absence of 10 μg of SEVI fibrils to give a final volume of 220 μl of solution. These incubations were performed in duplicate runs and allowed to equilibrate overnight at room temperature. After equilibration, each solution was centrifuged at 16,000×g for 30 min. The supernatants were separated from the pelleted fibrils, and 220 μl of fresh PBS was added to resuspend the pellets. A 100 μl aliquot of each resuspended pellet was pipetted into a cuvette (ultra-microcuvette, 10-mm light path, Hellma®, Müllheim, Germany), and the fluorescence of BTA-EG6 was determined at 355 nm excitation and 430 nm emission using a spectrofluorometer (Photon Technology International, Inc., Birmingham, N.J.). Error bars represent standard deviations from the mean. The graph was plotted and fitted using the following one-site specific binding algorithm to determine Kd: Y=Bmax×X/(Kd+X), where X is the concentration of BTA-EG6, Y is the specific binding fluorescence intensity, and Bmax corresponds to the apparent maximal observable fluorescence upon binding of BTA-EG6 to SEVI fibrils. The data were processed using Origin 7.0 (MicroCal Software, Inc., Northampton, Mass.).
  • Flow Cytometry
  • 105 Jurkat cells were incubated with biotinylated SEVI fibrils (40 μg/ml) with and without BTA-EG6 at a concentration of 10 (low) or 30 μg/ml (high) or heparin (100 μg/ml) as a positive control for interfering with SEVI binding to the cell surface. Cells were incubated for 1 h at 37° C., washed, and stained for 1 h with a covalent conjugate of streptavidin and fluorescein isothiocyanate (SA-FITC). Cells were washed and run on an Accuri C6 Flow Cytometer (Accuri Cytometers, Ann Arbor, Mich.). Data were analyzed using FlowJo (TreeStar Inc, Ashland, Oreg.).
  • Infectivity Assays
  • For infection of CEM-M7 cells, X4 tropic HIV-1IIIB (21 ng/ml p24) or R5 tropic HIV-1ADA (60 ng/ml p24) was pretreated for 10 min at room temperature with 15 μg/ml SEVI in the presence or absence of BTA-EG6. Treated virions were then added to 5×104 CEM-M7 cells/well in 96-well flat-bottomed tissue culture plates. After 2 h, the medium axis were replaced. Infection was assayed after 48 h by quantifying luciferase expression using the Promega Dual-Luciferase assay and a Beckman Coulter DTX880 plate reader.
  • For infections using semen, pooled human semen samples were added to virions at a 1:1 dilution and incubated for 15 min at room temperature in the presence or absence of BTA-EG6. After 15 min, the semen and virus mixture was diluted 1:15 into 5×104 CEM-M7 cells/well in a 96-well plate. Cells were washed after 1 h, and infection was assayed at 48 h as above.
  • For infections of PBMCs, R5 tropic HIV-1BaL preincubated with 15 μg/ml SEVI in the presence or absence of BTA-EG6 was added to 2×105 PHA/IL-2-stimulated PBMCs/well in 96-well flat-bottomed tissue culture plates. Cells were washed at 3 h, and infection was analyzed at day 4 using the HIV-1 p24 antigen capture assay (Advanced Bioscience Laboratory).
  • Virus Binding Assay
  • HIV-1 IIIB or ADA virions were pretreated with 15 μg/ml SEVI and added to 5×104 Jurkat cells, or A2En cells, in the presence or absence of BTA-EG6. After 90 min, cells were washed to remove any unbound virus, and bound virions were detected using an HIV-1 p24 antigen capture assay (Advanced Bioscience Laboratory).
  • Cytokine and Chemokine Studies
  • HIV-1 BaL virions were pretreated with 15 μg/ml SEVI and added to 5×104 A2En cells in the presence or absence of BTA-EG6. Supernatant was collected at 6 and 24 h, and the production of the chemokines IL-8 and Mip-3α was measured by ELISA (R&D Systems). To assess semen-mediated chemokine production, SiHa cells were treated with semen, as described above, in the presence or absence of BTA-EG6. After 6 h, supernatants were collected, and the production of the chemokines IL-8 and Mip-3α was measured by ELISA (R&D systems, Minneapolis, Minn.).
  • Toxicity Studies
  • The cervical epithelial cell lines SiHa, A2En (endocervical), and 3EC1 (ectocervical) were treated for 12 h with BTA-EG6 at concentrations up to 66 μg/ml, 10 times the IC50. After 12 h, cell viability was analyzed by measuring cellular metabolic activity using the resazurin cytotoxicity assay, AlamarBlue® (Invitrogen), in accordance with the manufacturer's protocol. Cytokine and chemokine production was assessed at 12 h by ELISA (R&D systems). Cells were also treated with 0.1% nonoxynol-9 as a positive control for cytotoxicity and with 0.1 μg/ml FSL1, a synthetic diacylated lipoprotein derived from Mycoplasma salivarium (InvivoGen, San Diego, Calif.), as a positive control for chemokine production.
  • SEVI fibrils are highly cationic, with a negative charge of +6.5 at neutral pH and +8 at pH 5, as would be seen in the vaginal mucosa. The cationic nature of SEVI is required for its ability to enhance HIV infection. This suggests that SEVI acts in a manner similar to other cationic polymers to enhance HIV infectivity.
  • To find compounds that bind SEVI fibrils and inhibit the ability of SEVI to enhance HIV infection, a model system of SEVI fibrils was developed. Fragments of prostatic acid phosphatase (PAP) from amino acid 248 to amino acid 286 were found to form SEVI fibrils. The PAP248-286 fragments at a concentration 10 mg/ml in PBS were agitated at 37° C. and 1400 RPM to form fibrils (FIG. 3A). The SEVI fibrils were viewed by electron microscopy at 72 hours (FIG. 3B).
  • To determine if the SEVI fibrils could enhance HIV-1 infection, CEM 5.25 cells were exposed to infectious HIV-1 for 2 hours in the presence or absence of SEVI. It was found that increasing concentrations of SEVI enhanced HIV-1 infection as evidenced by the increase in luciferase activity (FIG. 4A). Further, an increase in GFP expression indicative of HIV-1 infectivity was observed in cells treated with SEVI (FIG. 4B).
  • To determine if BTA-EG4 and BTA-EG6 were able to inhibit SEVI mediated enhancement of HIV infection, CEM 5.25 cells were exposed to infectious HIV-1 for 2 hours in the presence or absence of SEVI fibrils. In the presence of SEVI fibrils, increasing concentrations of BTA-EG4 and BTA-EG6 resulted in decreasing levels of luciferase activity (FIGS. 5A and 5B), indicating a decrease in the ability of SEVI fibrils to enhance HIV-1 infection.
  • To determine if BTA-EG4 and BTA-EG6 were capable of inhibiting semen mediated enhancement of HIV infection, HIV-1 IIIB virions were preincubated with 50% semen and increasing concentrations of BTA-EG4 and BTA-EG6. After 10 minutes, the stocks were diluted 15 fold and incubated with CEM 5.25 cells. The increasing concentrations of BTA-EG4 and BTA-EG6 resulted in a decrease in luciferase activity (FIGS. 6A and 6B), indicating that BTA-EG4 and BTA-EG6 were capable of inhibiting semen mediated enhancement of HIV infection. It was further found that BTA-EG4 and BTA-EG6 were capable of inhibiting SEVI-enhanced binding of HIV to the cell surface. HIV-1 IIIB virions were pretreated with 10 μg/mL SEVI and added to Jurkat cells with or without increasing concentrations of BTA-EG4 and BTA-EG6. After 90 minutes cells were washed to remove any unbound virus and bound virions were detected using a p24 ELISA. Increasing concentrations of BTA-EG4 and BTA-EG6 resulted in a decrease in HIV-1 binding to the cells (FIGS. 7A and 7B).
  • In order to find other small molecules that bind SEVI, a fluorescence polarizing screen was developed. The screen is shown in FIG. 8. Polarized light passed over a small unbound molecule with a fluorescent moiety will produce rapid rotation and will result in fluorescence that is de-polarized. Polarized light passed over SEVI bound to a small molecule with a fluorescent moiety will result in fluorescence that is polarized. As a proof of principle, SEVI fibrils were diluted to concentrations ranging from 5 to 100 μg/ml in the presence of 16 mg/ml of FITC-heparin. Samples were incubated at excitation of λ=480 and emission λ=535 (FIG. 8, left graph). When unlabeled heparin was added to the SEVI-FITC-heparin mixture, the polarization decreased as evidenced in FIG. 8, right graph. Using this assay, it was shown that BTA-EG4 and BTA-EG6 were also capable of binding SEVI fibrils. 100 mg/ml of SEVI was mixed with FITC-heparin in varying concentrations of BTA-EG4 and BTA-EG6. Samples were incubated for 1 hour at room temperature and polarized fluorescence was measured. It was shown that both BTA-EG4 and BTA-EG6 were capable of binding SEVI as evidenced by decreasing levels of polarized light (FIGS. 9A and 9B).
  • To determine if BTA-EG4 and BTA-EG6 were capable of being administered to cervical epithelial cells without cytotoxicity, the cervical cell lines A2En (endocervical) and SiHa cells were treated with BTA-EG4 and BTA-EG6 for 12 hours at concentrations up to 10 times greater than the inhibitory concentration. At 12 hours, viability was measured with Alamar Blue and it was shown that BTA-EG4 and BTA-EG6 did not affect the cell viability of A2En and SiHa cells. It was further shown in SiHa cells that BTA-EG4 and BTA-EG6 do not induce cytokine production. SiHa cells were treated with BTA-EG4 and BTA-EG6 at varying concentrations for 6 hours and cytokine production was examined by ELISA. As shown in FIGS. 11A-11C, IL-1b, MIP-3a, and TNF-a levels were unaffected by varying concentrations of BTA-EG4 and BTA-EG6.
  • The Thioflavin-T Analog BTA-EG6 Binds SEVI Fibrils
  • ThT is able to intercalate into the generic β-sheet structure of amyloid fibrils. The benzothiazole aniline derivative, BTA-EG6, is a ThT analog carrying a hexa(ethylene glycol) moiety (FIG. 12A). This molecule binds to Aβ fibrils and interferes with the ability of Aβ-binding proteins to interact with the fibrils. Fluorescence polarization was used to measure the ability of BTA-EG6 to bind SEVI. Increasing concentrations of BTA-EG6 were added to 50 μg/ml SEVI that had been preincubated with 16 μg/ml FITC-heparin, a known SEVI binder. BTA-EG6 was able to displace fluorescent heparin from the SEVI fibrils in a dose-dependent fashion (FIG. 12B), thus showing an interaction between these molecules and the fibrils.
  • Having observed an interaction between these molecules, the binding of BTA-EG6 to SEVI fibrils was assessed by quantifying its binding affinity. A fluorescence-based assay was used to determine the Kd between BTA-EG6 and the SEVI fibrils (see LeVine Amyloid 12(3): 12-15 (2005). FIG. 12C shows the relative fluorescence intensity (RFI) of BTA-EG6 bound to SEVI fibrils as a function of exposure of the SEVI peptides to increasing concentrations of BTA-EG6. Fitting the data in FIG. 12C with a one-site specific binding algorithm revealed a value of Kd=127±22 nM (R2=0.98). For comparison, this same fluorescence binding assay was used to measure the affinity of BTA-EG6 for binding to aggregated Alzheimer disease-related Aβ(1-42) peptides, which gave a value of Kd=111±32 nM (R2=0.95); this value was similar to the Kd value for binding of BTA-EG6 to SEVI fibrils.
  • To examine whether the interaction of BTA-EG6 with SEVI impacted the stability of the fibrils, preformed SEVI fibrils were incubated with BTA-EG6 for 3 h. After that time, fibrillar structures were examined by ThT fluorescence. ThT changes in fluorescence intensity when intercalated into the β-sheet structure common to amyloid fibrils; therefore, ThT fluorescence serves as a surrogate measure for fibrillar structure of SEVI and for the stability of SEVI fibrils. As seen in FIG. 12D, the addition of BTA-EG6 had no effect on fibrillar stability as measured by ThT. Unlike in the case with ThT, the fluorescence intensity of BTA-EG6 does not change upon binding to amyloid fibrils. The intrinsic fluorescence of BTA-EG6, therefore, does not interfere with the analysis of fibril stability using this assay. To further explore the interactions between SEVI fibrils and BTAEG6, the binding of this compound to SEVI was tested to determine if it could inhibit the ability of the fibrils to interact with the negatively charged surface of mammalian cells. Jurkat T-cells were incubated with 35 μg/ml SEVI-biotin fibrils, which were formed by fibrillization of a biotinylated PAP248-286 peptide, in the presence of 5.5 or 13 μg/ml BTA-EG6. Heparin was used as a positive control as this polyanionic compound has been shown to inhibit the binding of SEVI fibrils to the cell surface. Binding of the fibrils to the cell surface was detected using SA-FITC. As seen in FIG. 12E and Table 1, increasing concentrations of BTA-EG6 inhibited the ability of SEVI fibrils to interact with and bind the cell surface, as measured by both the percentage of cells with bound fibrils and the mean fluorescence intensity of the cells. Table 1 shows that BTA-EG6 binding to SEVI inhibits interaction of SEVI fibrils with the cell surface. Jurkat cells were incubated with SEVI-biotin (SEVI-Bio) for 1 h in the presence or absence of 5.5 (low) or 27 μg/ml (high) BTA-EG6. Surface-bound fibrils were detected with SA-FITC and measured by flow cytometry. Results are shown as percentage of cells with bound fibrils (SA-FITC+) as well as mean fluorescent intensity (MFI).
  • TABLE 1
    SA-FITC+
    Sample % MFI
    Unstained 1.65 28
    SEVI-Bio 48.1 2525
    SEVI-Bio + BTA-EG6 (low) 36.6 95.3
    SEVI-Bio + BTA-EG6 (high) 21.2 38.7
    SEVI-Bio + heparin 31.7 376
  • BTA-EG6 Inhibits SEVI-Mediated Enhancement of HIV-1 Infection
  • Having observed that BTA-EG6 was able to inhibit the interaction of SEVI with the cell surface, whether it could effectively inhibit SEVI-mediated enhancement of HIV-1 infection was investigated. CEM-M7 cells were infected with HIV-1 strain IIIB plus 15 μg/ml SEVI fibrils in the presence of increasing concentrations of BTA-EG6. CEM-M7 cells are a CD4+ CCR5+ CXCR4+T/B cell hybrid cell line and contain HIV LTR-driven luciferase and GFP reporter gene cassettes. The HIV LTR is a weak transcriptional regulator in the absence of its cognate, virally encoded trans-activator, Tat. As a result, luciferase and GFP expression levels in these cells are directly responsive to HIV-1 infection; this property therefore provides a convenient method to determine the extent of viral infection. BTA-EG6 was able to effectively inhibit SEVI-mediated enhancement of HIV infection in a dose-dependent fashion, reducing infectivity nearly back to baseline levels (i.e. levels detected in the absence of SEVI) at the highest concentrations tested (FIG. 13A). Importantly, BTA-EG6 had no effect on the infectivity of HIV virus alone, even at the highest concentrations (FIG. 13B), indicating that this effect was not due to direct inhibition of intrinsic virus infectivity.
  • Because most sexually transmitted HIV-1 infections are the result of R5 viruses, whether the effect of BTA-EG6 extended to a well characterized R5 strain was examined. CEM-M7 cells were infected with HIV-1ADA and 15 μg/ml SEVI, with and without increasing concentrations of BTA-EG6. Once again, BTA-EG6 showed a significant dose-dependent inhibition of SEVI-mediated enhancement of HIV-1 infection (FIG. 13C). No effect on the infectivity of HIV-1ADA was observed in the absence of SEVI (FIG. 13C). The IC50 of the BTA-EG6 for inhibition of SEVI-mediated enhancement of HIV-1 infection was also determined. To do this, CEM-M7 cells were infected with HIV-1ADA and 15 μg/ml SEVI in the presence of BTA-EG6. Ten different BTA-EG6 concentrations were tested, ranging from 0.4 to 50 μg/ml. The data were fit to an exponential decay curve to calculate the IC50, and results are shown in FIG. 13D. The calculated IC50 was 6.6 μg/ml for BTA-EG6 (equivalent to 13 μM).
  • Next, PBMCs were infected with HIV-1BAL with 15 μg/ml SEVI in the presence or absence of increasing concentrations of BTA-EG6. BTA-EG6 was able to inhibit SEVI-mediated enhancement of HIV-1 infection in PBMCs at similar concentrations to those seen in other cell lines (FIG. 13E). BTA-EG6 had no effect on the infectivity of HIV-1BaL in PBMCsin the absence of SEVI (FIG. 13E). Thus, the effects of BTA-EG6 are neither strain-dependent nor cell type-dependent, and the compound has no effect on HIV-1 infection in the absence of SEVI.
  • BTA-EG6 Inhibits Semen-Mediated Enhancement of HIV-1 Infection
  • For BTA-EG6 to be a viable microbicide candidate, it must be effective not just against the effects of SEVI but should be able to effectively inhibit the infection-enhancing activity of human semen. Therefore, the effect of this compound on semen-mediated enhancement of HIV-1 infection was examined. As human semen has been reported to be toxic to cultured cells, a protocol that minimized this toxicity was used. Pooled human semen was added to HIV-1IIIB virus in a 1:1 dilution. After 15 min, this solution was added to cells at a 1:15 dilution for a final concentration of 3.3%. As shown in FIG. 14A, BTA-EG6 efficiently inhibited the semen mediated enhancement of HIV-1 infection at similar concentrations to those active against SEVI alone. FIG. 14B shows that this effect extended to infection with an R5 virus, HIVADA as well.
  • A follow-up experiment was performed to test whether the effects of BTA-EG6 on semen were specific to the infection enhancing components in semen (i.e. SEVI) or due to a more general inhibitory effect against semen. To do this, BTA-EG6 inhibited semen-mediated chemokine release was examined. Human semen can be pro-inflammatory, mediating the release of IL-8 and MIP-3α from cervical endothelial cells. This property is thought to be due to the presence of several pro-inflammatory mediators but is not due to the presence of SEVI as SEVI does not stimulate the release of IL-8 or MIP-3α. SiHa cells, a cervical endothelial cell line, was treated with pooled human semen with or without 33 μg/ml BTA-EG6, a dose five times higher than the IC50. After 6 h, supernatants were collected and analyzed for production of IL-8 or MIP-3α. Pooled human semen effectively elicited the production of these chemokines from SiHa cells as expected, whereas BTA-EG6 had no inhibitory effect on semen-stimulated chemokine production (FIGS. 14C and D).
  • BTA-EG6 Inhibits SEVI-Mediated Attachment of HIV-1 to the Cell Surface
  • To more closely examine how BTA-EG6 mediates its inhibitory effects on SEVI-mediated HIV-1 infection enhancement, the ability of this compound to interfere with SEVI-enhanced binding of HIV-1 virions to the cell surface was examined. The cationic nature of SEVI enhances the binding of virions to the cell surface, which allows it to neutralize the electrostatic repulsion between the negatively charged HIV-1 virion and target cell surface. Jurkat T cells were incubated with HIV-1IIIB virions and 15 μg/ml SEVI in the presence or absence of increasing concentrations of BTA-EG6. Surface-bound virions were then measured by p24 ELISA after washing off unbound virus. SEVI was able to strongly enhance the binding of virions to the cell surface, and this effect was efficiently abrogated by BTAEG6 (FIG. 15A). BTA-EG6 had no effect on the binding of HIV virions to the cell surface in the absence of SEVI (FIG. 15A). Similar results were obtained with an R5 virus, HIV-1ADA (FIG. 4B). Additionally, this experiment was performed using A2En cells, a primary cell-derived endocervical cell line. It was found that SEVI also enhanced binding of virions to the surface of these cervical epithelial cells and that this effect was inhibited by BTA-EG6 (FIG. 15C).
  • Whether SEVI would increase HIV-1-mediated chemokine production and whether BTA-EG6 could inhibit this effect was also tested. HIV stimulates the release of MIP-3 and IL-8 from vaginal and cervical epithelial cells. Because SEVI increases the interactions between virions and epithelial cells, SEVI likely increases HIV-mediated chemokine release as well. Therefore, A2En cells were exposed to HIV-1BAL virions with and without SEVI, in the presence or absence of BTA-EG6. As seen in FIG. 4D, SEVI modestly increased the release of IL-8 from cells treated with virus, and BTA-EG6 was able to inhibit this release. Similar results were also obtained for MIP-3α.
  • BTA-EG6 is not Toxic to Cervical Cells
  • For a compound to be a legitimate HIV-1 microbicide candidate, it must not have toxic or inflammatory effects on the cervical endothelium. Loss of this protective layer leads to an increased ability for HIV-1 to cross the mucosal barrier, and inflammatory effects drive recruitment of HIV-1 target cells, further decreasing the natural barriers against successful transmission of HIV. Therefore, the effects of BTA-EG6 on cervical endothelial cells were examined. To do this, the following cell lines were used: 1) SiHa cells, a cervical carcinoma cell line; 2) A2En cells, a primary cell-derived line from the endocervical endothelium; and 3) 3EC1 cells, a primary cell-derived line from the ectocervical endothelium. To evaluate the effects of BTA-EG6 on cell viability, the compound was added to cells at concentrations up to 10× the IC50 for up to 24 h. Viability was assessed at 24 h by using the resazurin cytotoxicity assay. Resazurin cytotoxicity data were confirmed by trypan blue counts of viable cells. FIG. 16A shows that BTA-EG6 did not have any effects on cell viability, even at the highest concentrations tested.
  • Nonoxynol-9 (non-9), a spermicide, was used as a positive control. Whether treatment with BTA-EG6 led to the production of inflammatory cytokines and chemokines from the cervical cell lines was examined. All three cervical cell lines were treated for 6 h with concentrations of BTA-EG6 ranging from 6.6 to 66 μg/ml. Cell culture supernatants were then assessed for the presence of the inflammatory cytokines and chemokines Mip-3α (FIG. 5B), IL-8 (FIG. 16C), IL-1β, and TNF-α. These cytokines and chemokines were selected because they are up-regulated by other candidate microbicides and because they may play a role in microbicide-mediated enhancement of HIV-1 infection. BTA-EG6 did not lead to the release of any of these cytokines or chemokines, even at the highest doses tested. These results indicate that BTA-EG6 is not toxic to cervical endothelial cells.
  • BTA-EG6 inhibited SEVI-mediated enhancement of infection by both X4 (HIV-1IIIB) and R5 (HIV-1ADA) strains, in a dose-dependent fashion. In the case of HIV-1ADA, the IC50 was 13 μM; this value is 100-fold higher than the measured Kd of BTA-EG6 for binding to aggregated SEVI peptides (127 nM). Without being limited by theory, one explanation for this difference is that the ability of BTA-EG6 to compete with virion/fibril or virion/cell interactions requires a greater number of BTA-EG6 molecules than the noncompetitive binding of BTA-EG6 to SEVI alone. BTA-EG6 also inhibited SEVI-enhanced infection of primary cells (human peripheral blood mononuclear cells) in a dose-dependent fashion, and it blocked SEVI-enhanced binding of X4 (HIV-1IIIB) and R5 (HIV-1ADA) strains to target cells (including both Jurkat T cells and A2En endocervical cells). These data showed that (i) SEVI enhances the ability of HIV-1 virions to elicit IL-8 and MIP-3α from A2En endocervical cells and (ii) this can be inhibited by BTA-EG6. Without being limited by theory, these data show that BTA-EG6 and related compounds not only reduce the efficiency of HIV-1 infection of target cells but also reduce the level of target cell recruitment to virus-exposed genital mucosal tissue. BTA-EG6 effectively prevents semen mediated enhancement of HIV infectivity, showing that this activity of semen can be targeted by specifically inhibiting the SEVI fibrils. BTA-EG6 did not inhibit other properties of semen, such as the ability to elicit pro-inflammatory chemokines Thus, BTA-EG6 is an effective microbicide target
  • Example 3 Characterization of Monomeric and Oligomeric Binding to SEVI Fibrils
  • HIV-1 IIIB virions were pretreated with 15 μg/ml SEVI and added to 5×104 A2En cells (immortalized primary human endocervical cells) (FIG. 17A) or to Jurkat T cells (a CD4+ human T cell line) (FIG. 17B) in the presence or absence of test compound BTA-EG6 in monomeric, dimeric, trimeric, tetrameric or pentameric forms (at a final concentration of 25 μM). After 90 min, cells were washed to remove any unbound virus, and bound virions were detected using an HIV-1 p24 antigen capture assay (Advanced Bioscience Laboratory, Rockville, Md.). The data showed reduced HIV-1 p24 antigen capture in the presence of SEVI as compared to capture in the absence of SEVI.
  • These data suggest that the oligovalent scaffold may influence binding. FIG. 18 shows the structure of a benzothiazole aniline (BTA)-based monomer (1), dimer (2), trimer (3), tetramer (4), and pentamer (5) and a schematic of how the scaffold may affect binding. To test whether binding affinity/avidity is reduced in oligomeric forms, the binding of monomeric and oligomeric forms was assayed to determine the affinity/avidity constants (Kd) for the binding of monomer and oligomers 1-5 to amyloid fibrils formed from SEVI peptides. Affinity/avidity constants were estimated using a standard fluorescence assay, as described recently (J. S. Olsen et al, J. Biol. Chem., 2010, 285, 35488-35496). The BTA based monomer has a higher Kd than oligomeric forms. Estimated Kds are show in Table 2.
  • TABLE 2
    SEVI Fibrils
    Compound Kd (nM)
    Monomer (1) 107 ± 16 
    Dimer (2) 75 ± 10
    Trimer (3) 40 ± 6 
    Tetramer (4) 56 ± 6 
    Pentamer (5) 84 ± 21

Claims (47)

1. A method of treating or preventing a sexually transmitted infection in a subject, the method comprising administering to the subject a semen-derived enhancer of viral infection (SEVI)-binding agent, wherein the agent comprises a compound of the following formula:
Figure US20130157924A1-20130620-C00004
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
R1, R2, R3, R4, R5, R6, R7, and R8 are each independently selected from hydrogen, halogen, hydroxyl, trifluoromethyl, substituted or unsubstituted thio, substituted or unsubstituted alkoxyl, substituted or unsubstituted aryloxyl, substituted or unsubstituted amino, substituted or unsubstituted C1-12 alkyl, substituted or unsubstituted C2-12 alkenyl, substituted or unsubstituted C2-12 alkynyl, substituted or unsubstituted C1-12 heteroalkyl, substituted or unsubstituted C2-12 heteroalkenyl, substituted or unsubstituted C2-12 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and
R9 and R10 are each independently selected from hydrogen, substituted or unsubstituted C1-20 alkyl, substituted or unsubstituted C2-20 alkenyl, substituted or unsubstituted C2-20 alkynyl, substituted or unsubstituted C1-20 heteroalkyl, substituted or unsubstituted C2-20 heteroalkenyl, substituted or unsubstituted C2-20 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
2. The method of claim 1, wherein R3 is methyl.
3. The method of claim 1, wherein R1, R2, R4, R5, R6, R7, and R8 are hydrogen.
4. The method of claim 1, wherein R9 is
Figure US20130157924A1-20130620-C00005
wherein n is an integer from 0 to 20.
5. The method of claim 4, wherein n is 4 or 6.
6. (canceled)
7. The method of claim 1, wherein R10 is hydrogen.
8. The method of claim 1, wherein the compound is
Figure US20130157924A1-20130620-C00006
9. (canceled)
10. The method of claim 1, further comprising administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
11. The method of claim 1, wherein the sexually transmitted infection is selected from the group consisting of a viral infection, a bacterial infection, and a fungal infection.
12. (canceled)
13. (canceled)
14. (canceled)
15. A method of treating or preventing a sexually transmitted infection in a subject, the method comprising administering to the subject a semen-derived enhancer of viral infection (SEVI)-binding small molecule, wherein the SEVI-binding small molecule comprises a hydrophobic molecule, wherein the hydrophobic molecule incorporates into and binds the SEVI-fibrils.
16. The method of claim 15, further comprising administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
17. The method of claim 15, wherein the sexually transmitted infection is selected from the group consisting of a viral infection, a bacterial infection, and a fungal infection.
18. (canceled)
19. (canceled)
20. (canceled)
21. A method of treating or preventing a sexually transmitted infection in a subject, the method comprising administering to the subject a semen-derived enhancer of viral infection (SEVI)-binding small molecule, wherein the SEVI-binding small molecule comprises an anionic polypeptide supramolecular assembly.
22. The method of claim 21, further comprising administering to the subject an anti-viral, an anti-bacterial, or an anti-fungal agent.
23. The method of claim 21, wherein the sexually transmitted infection is selected from the group consisting of a viral infection, a bacterial infection, and a fungal infection.
24. (canceled)
25. (canceled)
26. (canceled)
27. The method of claim 21, wherein the anionic polypeptide supramolecular assembly is water-soluble.
28. The method of claim 21, wherein the anionic polypeptide supramolecular assembly comprises a soluble hydrogel and other supramolecular assemblies derived from an Ac-(XEXE)n-NH2 (SEQ ID NO:14) polypeptide and related polypeptides.
29. The method of claim 15, wherein the SEVI-binding small molecule further comprises a bulky side chain, a negatively charged side chain, a coupled moiety, and an antiviral molecule.
30. The method of claim 29, wherein the bulky side chain is poly-ethylene glycol.
31. The method of claim 29, wherein the antiviral molecule comprises pradimicin A or AZT.
32. A pharmaceutical composition comprising:
(a) a first agent, wherein the agent comprises a SEVI-binding agent comprising a compound of the following formula:
Figure US20130157924A1-20130620-C00007
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
R1, R2, R3, R4, R5, R6, R7, and R8 are each independently selected from hydrogen, halogen, hydroxyl, trifluoromethyl, substituted or unsubstituted thio, substituted or unsubstituted alkoxyl, substituted or unsubstituted aryloxyl, substituted or unsubstituted amino, substituted or unsubstituted C1-12 alkyl, substituted or unsubstituted C2-12 alkenyl, substituted or unsubstituted C2-12 alkynyl, substituted or unsubstituted C1-12 heteroalkyl, substituted or unsubstituted C2-12 heteroalkenyl, substituted or unsubstituted C2-12 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and
R9 and R10 are each independently selected from hydrogen, substituted or unsubstituted C1-20 alkyl, substituted or unsubstituted C2-20 alkenyl, substituted or unsubstituted C2-20 alkynyl, substituted or unsubstituted C1-20 heteroalkyl, substituted or unsubstituted C2-20 heteroalkenyl, substituted or unsubstituted C2-20 heteroalkynyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and
(b) a second agent selected from the group consisting of an anti-viral, an anti-bacterial, or an anti-fungal agent.
33. The pharmaceutical composition of claim 32, wherein R3 is methyl.
34. The pharmaceutical composition of claim 32, wherein R1, R2, R4, R5, R6, R7, and R8 are hydrogen.
35. The pharmaceutical composition of claim 32, wherein R9 is
Figure US20130157924A1-20130620-C00008
wherein n is an integer from 0 to 20.
36. The pharmaceutical composition of claim 35, wherein n is 4 or 6.
37. (canceled)
38. The pharmaceutical composition of claim 32, wherein R10 is hydrogen.
39. The pharmaceutical composition of claim 32, wherein the compound is
Figure US20130157924A1-20130620-C00009
40. (canceled)
41. A pharmaceutical composition comprising:
(a) a first agent, wherein the first agent comprises a semen-derived enhancer of viral infection (SEVI)-binding small molecule, wherein the SEVI-binding small molecule comprises a hydrophobic molecule, wherein the hydrophobic molecule incorporates into and binds the SEVI-fibrils.
(b) a second agent selected from the group consisting of an anti-viral, an anti-bacterial, and an anti-fungal agent.
42. A pharmaceutical composition comprising:
(a) a first agent, wherein the agent comprises a semen-derived enhancer of viral infection (SEVI)-binding small molecule, wherein the SEVI-binding small molecule comprises an anionic polypeptide supramolecular assembly.
(b) a second agent selected from the group consisting of an anti-viral, an anti-bacterial, and an anti-fungal agent.
43. The pharmaceutical composition of claim 42, wherein the anionic polypeptide supramolecular assembly is water-soluble.
44. The pharmaceutical composition of claim 42, wherein the anionic polypeptide supramolecular assembly comprises a soluble hydrogel and other supramolecular assemblies derived from an Ac-(XEXE)n-NH2 (SEQ ID NO:14) polypeptide and related polypeptides.
45. The pharmaceutical composition of claim 41, wherein the SEVI-binding small molecule further comprises a bulky side chain, a negatively charged side chain, a coupled moiety, and an antiviral molecule.
46. The pharmaceutical composition of claim 45, wherein the bulky side chain is poly-ethylene glycol (PEG).
47. The pharmaceutical composition of claim 45, wherein the antiviral molecule comprises pradimicin A or AZT.
US13/643,026 2010-04-23 2011-04-22 Reducing transmission of sexually transmitted infections Abandoned US20130157924A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/643,026 US20130157924A1 (en) 2010-04-23 2011-04-22 Reducing transmission of sexually transmitted infections

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US32746110P 2010-04-23 2010-04-23
PCT/US2011/033659 WO2011133929A2 (en) 2010-04-23 2011-04-22 Reducing transmission of sexually transmitted infections
US13/643,026 US20130157924A1 (en) 2010-04-23 2011-04-22 Reducing transmission of sexually transmitted infections

Publications (1)

Publication Number Publication Date
US20130157924A1 true US20130157924A1 (en) 2013-06-20

Family

ID=44834843

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/643,026 Abandoned US20130157924A1 (en) 2010-04-23 2011-04-22 Reducing transmission of sexually transmitted infections

Country Status (4)

Country Link
US (1) US20130157924A1 (en)
EP (1) EP2593103A4 (en)
CA (1) CA2797248A1 (en)
WO (1) WO2011133929A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8738188B2 (en) 2003-12-23 2014-05-27 Rain Bird Corporation Code replacement for irrigation controllers
US20160122401A1 (en) * 2013-04-30 2016-05-05 Forschungszentrum Juelich Gmbh Agents for preventing and treating hiv and other viral infections
US9348338B2 (en) 2003-12-23 2016-05-24 Rain Bird Corporation Modular and expandable irrigation controller
US20180289883A1 (en) * 2017-03-31 2018-10-11 InnAVasc Medical, Inc. Apparatus and method for cannulation of vascular access graft
US10582673B2 (en) 2002-10-15 2020-03-10 Rain Bird Corporation Modular and expandable irrigation controller
US11925782B2 (en) 2020-05-18 2024-03-12 InnAVasc Medical, Inc. Apparatus and method for cannulation of vascular access vessel

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013070910A1 (en) * 2011-11-08 2013-05-16 University Of Rochester Reducing transmission of sexually transmitted infections
CN108977412A (en) 2012-01-26 2018-12-11 生命科技公司 Method for improving viral infection
DE102013007405A1 (en) * 2013-04-30 2014-11-13 Forschungszentrum Jülich GmbH Agents for the prevention and treatment of HIV and other viral infections

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007011834A2 (en) * 2005-07-15 2007-01-25 The Regents Of The University Of California Compounds and method for the diagnosis and treatment of amyloid associated diseases

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002218897A1 (en) * 2000-12-18 2002-07-01 Boehringer Ingelheim (Canada) Ltd. Inhibitors of papilloma virus
EP1539151B1 (en) * 2002-08-02 2009-03-18 Genesoft Pharmaceuticals, Inc. Biaryl compounds having anti-infective activity
JP4206382B2 (en) * 2002-11-19 2009-01-07 アキリオン ファーマシューティカルズ,インコーポレーテッド Substituted arylthioureas and related compounds; inhibitors of viral replication
WO2006006172A2 (en) * 2004-07-15 2006-01-19 Ramot At Tel Aviv University Ltd. Use of anti-amyloid agents for treating and typing pathogen infections
WO2007002639A2 (en) * 2005-06-24 2007-01-04 Migenix Inc. Non-nucleoside anti-hepacivirus agents and uses thereof
BRPI0619733A2 (en) * 2005-12-12 2011-10-11 Genelabs Tech Inc n- (6-membered aromatic ring) -amido antiviral compounds
US8183236B2 (en) * 2007-04-12 2012-05-22 University Of Southern California Compounds with HIV-1 integrase inhibitory activity and use thereof as anti-HIV/AIDS therapeutics

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007011834A2 (en) * 2005-07-15 2007-01-25 The Regents Of The University Of California Compounds and method for the diagnosis and treatment of amyloid associated diseases

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
Easterhoff, David et al; "Semen derived enhancer of viral infection (SEVI) binds bacteria, enhances bacterial phagocytosis by macrophages, and can protect against vaginal infection by a sexually transmited bacterial pathogen." Antimicrob. Agent. Chemother. (2013) 57(6) p2443-2450 *
Entry for the American journal of syphilis, gonorrhea and venerial diseases in the library of the University of Toronto, downloaded 19 Feb 2014 *
Fauci, Anthony S.; "Pathogeneisis of hiv disease: opportunities for new prevention intervensions." Clin. Infect. Disease (2007) 45 pS206-212 *
Fauci, Anthony S.; "Pathogenesis of HIV disease: opportunities for new prevention interventions." Clin. Infect. Disease (2007) 45 pS206-212 *
Hong, Seunghee et al; "Fibrils of prostatic acid phosphatase fragments boost infections with xmrv(xenotoropic murine leukemia virus related virus), a human retrovirus associated with prostate cancer." J. Virology (2009) 83(14) p6995-7003 *
Kenakin, Terry; "Receptor Theory." Curr. Protoc. Pharmacol. (2008) 41, p1.2.1-1.2.28 *
Levine, Harry III; "Multiple ligand binding sites on abeta(1-40) fibrils." Amyloid (2005) 12(1) p5-14) *
Mase, Toshiaki and Itoh, Takahiro; "General and practical synthesis of benzothiazoles." Pure App. Chem. (2008) 80(4) p707-715 *
Mayer, Kenneth H. et al; "Safety and tolerability of vaginal pro 2000 gel in sexually active hiv-uninfected and abstinent hiv-infected women." AIDS (2003) 17 p321-329 *
Münch, Jan et al; "Semen derived amyloid fibrils drastically enhance HIV infection." Cell (2007) 131 p1059-1071 *
Price, Richard W.; "Neurological complications of hiv infection." Lancet (1996) 348 p445-452 *
Roan, Nadia R. et al; "Aminoquinoline surfen inhibits the action of sevi(semen derived enhancer of viral infection)." J. Biol. Chem. (2010) 285(3) p1861-1869, published online 6 Nov 2009 *
Roan, Nadia R. et al; "Aminoquinoline surfen inhyibits the action of SEVI (semen derived enhancer of viral infection)." J. Biol. Chem. (2010) 285(3) p1861-1869, published online 6 Nov 2009 *
Wong, Yue-Ling et al; "A structure activity study of spermicidal and anti-hiv properties of hydroxylated cationic surfactants." Bioorg. Med. Chem. (2002) 10 p3599-3608 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10582673B2 (en) 2002-10-15 2020-03-10 Rain Bird Corporation Modular and expandable irrigation controller
US11559013B2 (en) 2002-10-15 2023-01-24 Rain Bird Corporation Expandable irrigation controller
US11071263B2 (en) 2002-10-15 2021-07-27 Rain Bird Corporation Expandable irrigation controller
US10663941B2 (en) 2003-12-23 2020-05-26 Rain Bird Corporation Code replacement for irrigation controllers
US9348338B2 (en) 2003-12-23 2016-05-24 Rain Bird Corporation Modular and expandable irrigation controller
US11096341B2 (en) 2003-12-23 2021-08-24 Rain Bird Corporation Modular and expandable irrigation controller
US10025284B2 (en) 2003-12-23 2018-07-17 Rain Bird Corporation Code replacement for irrigation controllers
US9547313B2 (en) 2003-12-23 2017-01-17 Rain Bird Corporation Code replacement for irrigation controllers
US8738188B2 (en) 2003-12-23 2014-05-27 Rain Bird Corporation Code replacement for irrigation controllers
US11163284B2 (en) 2007-06-22 2021-11-02 Rain Bird Corporation Code replacement for irrigation controllers
US11630431B2 (en) 2007-06-22 2023-04-18 Rain Bird Corporation Code replacement for irrigation controllers
US10030056B2 (en) * 2013-04-30 2018-07-24 Forschungszentrum Juelich Gmbh Agents for preventing and treating HIV and other viral infections
US20160122401A1 (en) * 2013-04-30 2016-05-05 Forschungszentrum Juelich Gmbh Agents for preventing and treating hiv and other viral infections
US11065377B2 (en) * 2017-03-31 2021-07-20 InnAVasc Medical, Inc. Apparatus and method for cannulation of vascular access graft
US20180289883A1 (en) * 2017-03-31 2018-10-11 InnAVasc Medical, Inc. Apparatus and method for cannulation of vascular access graft
US11925781B2 (en) 2018-10-30 2024-03-12 InnAVasc Medical, Inc. Apparatus and method for cannulation of vascular access vessel
US11925782B2 (en) 2020-05-18 2024-03-12 InnAVasc Medical, Inc. Apparatus and method for cannulation of vascular access vessel

Also Published As

Publication number Publication date
CA2797248A1 (en) 2011-10-27
WO2011133929A2 (en) 2011-10-27
WO2011133929A3 (en) 2012-04-05
EP2593103A2 (en) 2013-05-22
EP2593103A4 (en) 2014-02-19

Similar Documents

Publication Publication Date Title
US20130157924A1 (en) Reducing transmission of sexually transmitted infections
Veazey et al. Protection of macaques from vaginal SHIV challenge by vaginally delivered inhibitors of virus–cell fusion
Olsen et al. Amyloid-binding small molecules efficiently block SEVI (semen-derived enhancer of virus infection)-and semen-mediated enhancement of HIV-1 infection
JP4413781B2 (en) Non-nucleoside reverse transcriptase inhibitors
US20090176776A1 (en) Small molecule inhibitors of hiv-1 capsid assembly
US20030225155A1 (en) Pharmacological agents and methods of treatment that inactivate pathogenic prokaryotic and eukaryotic cells and viruses by attacking highly conserved domains in structural metalloprotein and metalloenzyme targets
US8546439B2 (en) Small molecule inhibitors of retroviral assembly and maturation
JP2002540156A (en) Thiadiazolyl urea or thiourea derivatives for antiviral therapy
JP5372751B2 (en) AZA-peptide protease inhibitor
CA2818308A1 (en) Bifunctional molecules with antibody-recruiting and entry inhibitory activity against the human immunodeficiency virus
US9610264B2 (en) Compounds for the treatment and prevention of retroviral infections
KR20010071580A (en) Compounds obtained from salvia species having antiviral activity
US20090048222A1 (en) Triaza Compound Immunoregulatory Agents
CA2001359C (en) Methods for the treatment and prophylaxis of pneumocystis carinii pneumonia and other diseases and compounds and formulations for use in said methods
JPH09508369A (en) Method for inhibiting retrovirus integrase
Rommasi et al. Antiviral drugs proposed for COVID-19: action mechanism and pharmacological data.
US6342492B1 (en) Anti-viral triaza compounds and compositions
JP2009242247A (en) Human immunodeficiency virus infection inhibitor and aids-treating or preventing drug
EA010483B1 (en) Agent for inhibiting membrane virus production, method for the production thereof, pharmaceutical composition and method for inhibiting viral infections
US20140323420A1 (en) Reducing transmission of sexually transmitted infections
US9333183B2 (en) Self-assembling polyphenol-quinonoid polymer derivatives and uses thereof
JP2002540150A (en) Virus treatment
AU2015214176A1 (en) Treating Flavivirus infections with amodiaquine and derivatives thereof
FR2722692A1 (en) Medicament for treating retrovirus infections contg. phospholipase A2 from bee venom
Kumar Design, Synthesis and Biological Evaluation of Benzpyridine Derivatives As Anti-HIV agents

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF ROCHESTER;REEL/FRAME:030920/0211

Effective date: 20130729

AS Assignment

Owner name: UNIVERSITY OF ROCHESTER, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DEWHURST, STEPHEN;NILSSON, BRADLEY;OLSEN, JOANNA;REEL/FRAME:032312/0678

Effective date: 20110728

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:YANG, JERRY;REEL/FRAME:032312/0743

Effective date: 20110727

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF ROCHESTER;REEL/FRAME:038243/0694

Effective date: 20160411